Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Neuron ; 111(22): 3619-3633.e8, 2023 11 15.
Article in English | MEDLINE | ID: mdl-37689059

ABSTRACT

A pathological hallmark of Alzheimer's disease (AD) is the deposition of amyloid-ß (Aß) protein in the brain. Physical exercise has been shown to reduce Aß burden in various AD mouse models, but the underlying mechanisms have not been elucidated. Irisin, an exercise-induced hormone, is the secreted form of fibronectin type-III-domain-containing 5 (FNDC5). Here, using a three-dimensional (3D) cell culture model of AD, we show that irisin significantly reduces Aß pathology by increasing astrocytic release of the Aß-degrading enzyme neprilysin (NEP). This is mediated by downregulation of ERK-STAT3 signaling. Finally, we show that integrin αV/ß5 acts as the irisin receptor on astrocytes required for irisin-induced release of astrocytic NEP, leading to clearance of Aß. Our findings reveal for the first time a cellular and molecular mechanism by which exercise-induced irisin attenuates Aß pathology, suggesting a new target pathway for therapies aimed at the prevention and treatment of AD.


Subject(s)
Alzheimer Disease , Neprilysin , Mice , Animals , Neprilysin/genetics , Neprilysin/metabolism , Fibronectins/metabolism , Down-Regulation , Astrocytes/metabolism , Amyloid beta-Peptides/metabolism , Alzheimer Disease/metabolism , Brain/metabolism
2.
Front Pharmacol ; 14: 1190402, 2023.
Article in English | MEDLINE | ID: mdl-37601062

ABSTRACT

Therapeutics discovery and development for Alzheimer's disease (AD) has been an area of intense research to alleviate memory loss and the underlying pathogenic processes. Recent drug discovery approaches have utilized in silico computational strategies for drug candidate selection which has opened the door to repurposing drugs for AD. Computational analysis of gene expression signatures of patients stratified by the APOE4 risk allele of AD led to the discovery of the FDA-approved drug bumetanide as a top candidate agent that reverses APOE4 transcriptomic brain signatures and improves memory deficits in APOE4 animal models of AD. Bumetanide is a loop diuretic which inhibits the kidney Na+-K+-2Cl- cotransporter isoform, NKCC2, for the treatment of hypertension and edema in cardiovascular, liver, and renal disease. Electronic health record data revealed that patients exposed to bumetanide have lower incidences of AD by 35%-70%. In the brain, bumetanide has been proposed to antagonize the NKCC1 isoform which mediates cellular uptake of chloride ions. Blocking neuronal NKCC1 leads to a decrease in intracellular chloride and thus promotes GABAergic receptor mediated hyperpolarization, which may ameliorate disease conditions associated with GABAergic-mediated depolarization. NKCC1 is expressed in neurons and in all brain cells including glia (oligodendrocytes, microglia, and astrocytes) and the vasculature. In consideration of bumetanide as a repurposed drug for AD, this review evaluates its pharmaceutical properties with respect to its estimated brain levels across doses that can improve neurologic disease deficits of animal models to distinguish between NKCC1 and non-NKCC1 mechanisms. The available data indicate that bumetanide efficacy may occur at brain drug levels that are below those required for inhibition of the NKCC1 transporter which implicates non-NKCC1 brain mechansims for improvement of brain dysfunctions and memory deficits. Alternatively, peripheral bumetanide mechanisms may involve cells outside the central nervous system (e.g., in epithelia and the immune system). Clinical bumetanide doses for improved neurological deficits are reviewed. Regardless of mechanism, the efficacy of bumetanide to improve memory deficits in the APOE4 model of AD and its potential to reduce the incidence of AD provide support for clinical investigation of bumetanide as a repurposed AD therapeutic agent.

3.
Phytomedicine ; 104: 154158, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35728383

ABSTRACT

BACKGROUND: Based on the complex pathology of AD, a single chemical approach may not be sufficient to deal simultaneously with multiple pathways of amyloid-tau neuroinflammation. A polydrug approach which contains multiple bioactive components targeting multiple pathways in AD would be more appropriate. Here we focused on a Chinese medicine (HLXL), which contains 56 bioactive natural products identified in 11 medicinal plants and displays potent anti-inflammatory and immuno-modulatory activity. HYPOTHESIS/PURPOSE: We investigated the neuroimmune and neuroinflammation mechanisms by which HLXL may attenuate AD neuropathology. Specifically, we investigated the effects of HLXL on the neuropathology of AD using both transgenic mouse models as well as microglial cell-based models. STUDY DESIGN: The 5XFAD transgenic animals and microglial cell models were respectively treated with HLXL and Aß42, and/or lipopolysaccharide (LPS), and then analyzed focusing on microglia mediated Aß uptake and clearance, as well as pathway changes. METHODS: We showed that HLXL significantly reduced amyloid neuropathology by upregulation of microglia-mediated phagocytosis of Aß both in vivo and in vitro. HLXL displayed multi-modal mechanisms regulating pathways of phagocytosis and energy metabolism. RESULTS: Our results may not only open a new avenue to support pharmacologic modulation of neuroinflammation and the neuroimmune system for AD intervention, but also identify HLXL as a promising natural medicine for AD. CONCLUSION: It is conceivable that the traditional wisdom of natural medicine in combination with modern science and technology would be the best strategy in developing effective therapeutics for AD.


Subject(s)
Alzheimer Disease , Amyloidosis , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Disease Models, Animal , Mice , Mice, Transgenic , Microglia , Neuroinflammatory Diseases , Phagocytosis
4.
J Alzheimers Dis ; 80(2): 775-786, 2021.
Article in English | MEDLINE | ID: mdl-33579853

ABSTRACT

BACKGROUND: Cromolyn is an anti-neuroinflammatory modulator with a multifactorial mechanism of action that has been shown to inhibit amyloid-ß (Aß) aggregation and enhance microglial uptake and clearance of Aß. OBJECTIVE: We report the effects of fluoro-cromolyn derivatives on microglial cell toxicity and microglial clearance of Aß42. METHODS: Microglial cell toxicity for cromolyn derivatives were determined in naive BV2 microglial cells. Microglial clearance assays were performed with Aß42 in naive BV2 microglial cell line and single cell clone BV2 line expressing CD33WT. PET imaging was performed for three F-18 analogs in a rhesus macaque. RESULTS: All compounds but derivative 8 exhibited low microglial cell toxicity. Cromolyn 1 and derivatives 2, 4, and 7 displayed an increased uptake on Aß42 in naïve BV2 microglial cells. Derivative 4 increased Aß42 uptake in a dose-dependent manner and at 75µM resulted in a one-fold increase in Aß42 uptake in BV2-CD33WT. PET imaging for three [18F]cromolyn analogs revealed the order of brain tracer penetration to be 4a > 10 > 2a. Tracer 4a exhibited enhanced uptake in areas of high perfusion (putamen, grey matter, and cerebellum) and lower signal in areas of lower perfusion (caudate, thalamus, and white matter). CONCLUSION: Substantial uptake of Aß42 in both naïve BV2 and BV2-CD33WT cells observed with 4 indicate conversion of microglial cells from a pro-inflammatory to an activation state favoring Aß phagocytosis/clearance. These findings suggest that a fluoro-cromolyn analog could reduce fibril-prone Aß42in vivo and thereby serve as a therapeutic for the treatment and prevention of AD.


Subject(s)
Alzheimer Disease/drug therapy , Cromolyn Sodium/pharmacology , Microglia/drug effects , Microglia/metabolism , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Brain/drug effects , Brain/metabolism , Cell Line , Cells, Cultured , Cromolyn Sodium/metabolism , Macaca mulatta/metabolism , Mice , Neuroprotective Agents/pharmacology , Peptide Fragments/metabolism , Phagocytosis/drug effects
5.
Sci Rep ; 8(1): 1144, 2018 01 18.
Article in English | MEDLINE | ID: mdl-29348604

ABSTRACT

Amyloid-beta protein (Aß) deposition is a pathological hallmark of Alzheimer's disease (AD). Aß deposition triggers both pro-neuroinflammatory microglial activation and neurofibrillary tangle formation. Cromolyn sodium is an asthma therapeutic agent previously shown to reduce Aß levels in transgenic AD mouse brains after one-week of treatment. Here, we further explored these effects as well as the mechanism of action of cromolyn, alone, and in combination with ibuprofen in APPSwedish-expressing Tg2576 mice. Mice were treated for 3 months starting at 5 months of age, when the earliest stages of ß-amyloid deposition begin. Cromolyn, alone, or in combination with ibuprofen, almost completely abolished longer insoluble Aß species, i.e. Aß40 and Aß42, but increased insoluble Aß38 levels. In addition to its anti-aggregation effects on Aß, cromolyn, alone, or plus ibuprofen, but not ibuprofen alone, increased microglial recruitment to, and phagocytosis of ß-amyloid deposits in AD mice. Cromolyn also promoted Aß42 uptake in microglial cell-based assays. Collectively, our data reveal robust effects of cromolyn, alone, or in combination with ibuprofen, in reducing aggregation-prone Aß levels and inducing a neuroprotective microglial activation state favoring Aß phagocytosis versus a pro-neuroinflammatory state. These findings support the use of cromolyn, alone, or with ibuprofen, as a potential AD therapeutic.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Protein Precursor/antagonists & inhibitors , Cromolyn Sodium/pharmacology , Ibuprofen/pharmacology , Microglia/drug effects , Neuroprotective Agents/pharmacology , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/biosynthesis , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Anti-Asthmatic Agents/pharmacology , Brain/drug effects , Brain/metabolism , Brain/pathology , Disease Models, Animal , Drug Combinations , Drug Repositioning , Gene Expression , Humans , Male , Mice , Mice, Transgenic , Microglia/cytology , Microglia/metabolism , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/biosynthesis , Phagocytosis/drug effects , Transgenes
6.
Proc Natl Acad Sci U S A ; 114(23): E4676-E4685, 2017 06 06.
Article in English | MEDLINE | ID: mdl-28533375

ABSTRACT

The activity of the transcription factor nuclear factor-erythroid 2 p45-derived factor 2 (NRF2) is orchestrated and amplified through enhanced transcription of antioxidant and antiinflammatory target genes. The present study has characterized a triazole-containing inducer of NRF2 and elucidated the mechanism by which this molecule activates NRF2 signaling. In a highly selective manner, the compound covalently modifies a critical stress-sensor cysteine (C151) of the E3 ligase substrate adaptor protein Kelch-like ECH-associated protein 1 (KEAP1), the primary negative regulator of NRF2. We further used this inducer to probe the functional consequences of selective activation of NRF2 signaling in Huntington's disease (HD) mouse and human model systems. Surprisingly, we discovered a muted NRF2 activation response in human HD neural stem cells, which was restored by genetic correction of the disease-causing mutation. In contrast, selective activation of NRF2 signaling potently repressed the release of the proinflammatory cytokine IL-6 in primary mouse HD and WT microglia and astrocytes. Moreover, in primary monocytes from HD patients and healthy subjects, NRF2 induction repressed expression of the proinflammatory cytokines IL-1, IL-6, IL-8, and TNFα. Together, our results demonstrate a multifaceted protective potential of NRF2 signaling in key cell types relevant to HD pathology.


Subject(s)
Huntington Disease/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , NF-E2-Related Factor 2/metabolism , Adult , Aged , Animals , Brain/drug effects , Brain/metabolism , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Female , HEK293 Cells , Humans , Huntington Disease/genetics , Kelch-Like ECH-Associated Protein 1/chemistry , MPTP Poisoning/metabolism , MPTP Poisoning/prevention & control , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Middle Aged , NF-E2-Related Factor 2/chemistry , Neural Stem Cells/metabolism , Neuroprotective Agents/pharmacology , Protein Conformation/drug effects , Rats , Signal Transduction
7.
J Pharmacol Exp Ther ; 362(1): 31-44, 2017 07.
Article in English | MEDLINE | ID: mdl-28416568

ABSTRACT

Alzheimer's disease (AD) is characterized neuropathologically by an abundance of 1) neuritic plaques, which are primarily composed of a fibrillar 42-amino-acid amyloid-ß peptide (Aß), as well as 2) neurofibrillary tangles composed of aggregates of hyperphosporylated tau. Elevations in the concentrations of the Aß42 peptide in the brain, as a result of either increased production or decreased clearance, are postulated to initiate and drive the AD pathologic process. We initially introduced a novel class of bridged aromatics referred tγ-secretase modulatoro as γ-secretase modulators that inhibited the production of the Aß42 peptide and to a lesser degree the Aß40 peptide while concomitantly increasing the production of the carboxyl-truncated Aß38 and Aß37 peptides. These modulators potently lower Aß42 levels without inhibiting the γ-secretase-mediated proteolysis of Notch or causing accumulation of carboxyl-terminal fragments of APP. In this study, we report a large number of pharmacological studies and early assessment of toxicology characterizing a highly potent γ-secretase modulator (GSM), (S)-N-(1-(4-fluorophenyl)ethyl)-6-(6-methoxy-5-(4-methyl-1H-imidazol-1-yl)pyridin-2-yl)-4-methylpyridazin-3-amine (BPN-15606). BPN-15606 displayed the ability to significantly lower Aß42 levels in the central nervous system of rats and mice at doses as low as 5-10 mg/kg, significantly reduce Aß neuritic plaque load in an AD transgenic mouse model, and significantly reduce levels of insoluble Aß42 and pThr181 tau in a three-dimensional human neural cell culture model. Results from repeat-dose toxicity studies in rats and dose escalation/repeat-dose toxicity studies in nonhuman primates have designated this GSM for 28-day Investigational New Drug-enabling good laboratory practice studies and positioned it as a candidate for human clinical trials.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid beta-Peptides/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/toxicity , Peptide Fragments/antagonists & inhibitors , Phenethylamines/pharmacology , Phenethylamines/toxicity , Pyridazines/pharmacology , Pyridazines/toxicity , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Cells, Cultured , Central Nervous System/drug effects , Central Nervous System/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacokinetics , Humans , Macaca fascicularis , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Peptide Fragments/genetics , Peptide Fragments/metabolism , Plaque, Amyloid/drug therapy , Rats , Rats, Sprague-Dawley , tau Proteins/metabolism
8.
J Huntingtons Dis ; 5(4): 347-355, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27983565

ABSTRACT

BACKGROUND: Modulation of gene transcription by HDAC inhibitors has been shown repeatedly to be neuroprotective in cellular, invertebrate, and rodent models of Huntington's disease (HD). It has been difficult to translate these treatments to the clinic, however, because existing compounds have limited potency or brain bioavailability. OBJECTIVE: In the present study, we assessed the therapeutic potential of LBH589, an orally bioavailable hydroxamic acid-derived nonselective HDAC inhibitor in mouse models of HD. METHOD: The efficacy of LBH589 is tested in two HD mouse models using various biochemical, behavioral and neuropathological outcome measures. RESULTS: We show that LBH589 crosses the blood brain barrier; induces histone hyperacetylation and prevents striatal neuronal shrinkage in R6/2 HD mice. In full-length knock-in HD mice LBH589-treatment improves motor performance and reduces neuronal atrophy. CONCLUSIONS: Our efficacious results of LBH589 in fragment and full-length mouse models of HD suggest that LBH589 is a promising candidate for clinical assessment in HD patients and provides confirmation that non-selective HDAC inhibitors can be viable clinical candidates.


Subject(s)
Histone Deacetylase Inhibitors/pharmacology , Huntington Disease/drug therapy , Hydroxamic Acids/pharmacology , Indoles/pharmacology , Neuroprotective Agents/pharmacology , Animals , Atrophy/drug therapy , Atrophy/metabolism , Atrophy/pathology , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Corpus Striatum/pathology , Disease Models, Animal , Female , Gene Knock-In Techniques , Histone Deacetylase Inhibitors/pharmacokinetics , Histones/metabolism , Huntington Disease/pathology , Huntington Disease/physiopathology , Hydroxamic Acids/pharmacokinetics , Indoles/pharmacokinetics , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Motor Activity/drug effects , Neurons/drug effects , Neurons/pathology , Neurons/physiology , Neuroprotective Agents/pharmacokinetics , Panobinostat
9.
Mol Neurodegener ; 11(1): 75, 2016 12 09.
Article in English | MEDLINE | ID: mdl-27938410

ABSTRACT

Alzheimer's disease (AD) transgenic mice have been used as a standard AD model for basic mechanistic studies and drug discovery. These mouse models showed symbolic AD pathologies including ß-amyloid (Aß) plaques, gliosis and memory deficits but failed to fully recapitulate AD pathogenic cascades including robust phospho tau (p-tau) accumulation, clear neurofibrillary tangles (NFTs) and neurodegeneration, solely driven by familial AD (FAD) mutation(s). Recent advances in human stem cell and three-dimensional (3D) culture technologies made it possible to generate novel 3D neural cell culture models that recapitulate AD pathologies including robust Aß deposition and Aß-driven NFT-like tau pathology. These new 3D human cell culture models of AD hold a promise for a novel platform that can be used for mechanism studies in human brain-like environment and high-throughput drug screening (HTS). In this review, we will summarize the current progress in recapitulating AD pathogenic cascades in human neural cell culture models using AD patient-derived induced pluripotent stem cells (iPSCs) or genetically modified human stem cell lines. We will also explain how new 3D culture technologies were applied to accelerate Aß and p-tau pathologies in human neural cell cultures, as compared the standard two-dimensional (2D) culture conditions. Finally, we will discuss a potential impact of the human 3D human neural cell culture models on the AD drug-development process. These revolutionary 3D culture models of AD will contribute to accelerate the discovery of novel AD drugs.


Subject(s)
Alzheimer Disease , Cell Culture Techniques , Neural Stem Cells/cytology , Humans
10.
Cell Chem Biol ; 23(7): 849-861, 2016 07 21.
Article in English | MEDLINE | ID: mdl-27427231

ABSTRACT

There are currently no disease-modifying therapies for the neurodegenerative disorder Huntington's disease (HD). This study identified novel thiazole-containing inhibitors of the deacetylase sirtuin-2 (SIRT2) with neuroprotective activity in ex vivo brain slice and Drosophila models of HD. A systems biology approach revealed an additional SIRT2-independent property of the lead-compound, MIND4, as an inducer of cytoprotective NRF2 (nuclear factor-erythroid 2 p45-derived factor 2) activity. Structure-activity relationship studies further identified a potent NRF2 activator (MIND4-17) lacking SIRT2 inhibitory activity. MIND compounds induced NRF2 activation responses in neuronal and non-neuronal cells and reduced production of reactive oxygen species and nitrogen intermediates. These drug-like thiazole-containing compounds represent an exciting opportunity for development of multi-targeted agents with potentially synergistic therapeutic benefits in HD and related disorders.


Subject(s)
Disease Models, Animal , Huntington Disease/drug therapy , NF-E2-Related Factor 2/antagonists & inhibitors , Neuroprotective Agents/pharmacology , Sirtuin 2/antagonists & inhibitors , Thiazoles/pharmacology , Thiazoles/therapeutic use , Animals , Cell Line , Dose-Response Relationship, Drug , Drosophila , Huntington Disease/metabolism , NF-E2-Related Factor 2/metabolism , Neuroprotective Agents/chemistry , Neuroprotective Agents/therapeutic use , Rats , Sirtuin 2/deficiency , Sirtuin 2/metabolism , Structure-Activity Relationship , Thiazoles/chemistry
11.
ACS Med Chem Lett ; 6(5): 607-11, 2015 May 14.
Article in English | MEDLINE | ID: mdl-26005542

ABSTRACT

Inhibitors of sirtuin-2 (SIRT2) deacetylase have been shown to be protective in various models of Huntington's disease (HD) by decreasing polyglutamine aggregation, a hallmark of HD pathology. The present study was directed at optimizing the potency of SIRT2 inhibitors containing the 3-(benzylsulfonamido)benzamide scaffold and improving their metabolic stability. Molecular modeling and docking studies revealed an unfavorable role of the sulfonamide moiety for SIRT2 binding. This prompted us to replace the sulfonamide with thioether, sulfoxide, or sulfone groups. The thioether analogues were the most potent SIRT2 inhibitors with a two- to three-fold increase in potency relative to their corresponding sulfonamide analogues. The newly synthesized compounds also demonstrated higher SIRT2 selectivity over SIRT1 and SIRT3. Two thioether-derived compounds (17 and 18) increased α-tubulin acetylation in a dose-dependent manner in at least one neuronal cell line, and 18 was found to inhibit polyglutamine aggregation in PC12 cells.

12.
PLoS One ; 10(1): e0116919, 2015.
Article in English | MEDLINE | ID: mdl-25608039

ABSTRACT

Sirtuin deacetylases regulate diverse cellular pathways and influence disease processes. Our previous studies identified the brain-enriched sirtuin-2 (SIRT2) deacetylase as a potential drug target to counteract neurodegeneration. In the present study, we characterize SIRT2 inhibition activity of the brain-permeable compound AK7 and examine the efficacy of this small molecule in models of Parkinson's disease, amyotrophic lateral sclerosis and cerebral ischemia. Our results demonstrate that AK7 is neuroprotective in models of Parkinson's disease; it ameliorates alpha-synuclein toxicity in vitro and prevents 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced dopamine depletion and dopaminergic neuron loss in vivo. The compound does not show beneficial effects in mouse models of amyotrophic lateral sclerosis and cerebral ischemia. These findings underscore the specificity of protective effects observed here in models of Parkinson's disease, and previously in Huntington's disease, and support the development of SIRT2 inhibitors as potential therapeutics for the two neurodegenerative diseases.


Subject(s)
Amyotrophic Lateral Sclerosis/physiopathology , Benzamides/administration & dosage , Brain Ischemia/physiopathology , Neuroprotective Agents/administration & dosage , Parkinson Disease/prevention & control , Sirtuin 2/antagonists & inhibitors , Small Molecule Libraries/administration & dosage , Sulfonamides/administration & dosage , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/adverse effects , Animals , Benzamides/pharmacology , Cell Line , Disease Models, Animal , Humans , Male , Mice , Neuroprotective Agents/pharmacology , Parkinson Disease/metabolism , Small Molecule Libraries/pharmacology , Sulfonamides/pharmacology , alpha-Synuclein/metabolism
13.
Eur J Med Chem ; 76: 414-26, 2014 Apr 09.
Article in English | MEDLINE | ID: mdl-24602787

ABSTRACT

Inhibitors of sirtuin-2 deacetylase (SIRT2) have been shown to be protective in various models of Huntington's disease (HD) by decreasing polyglutamine aggregation, a hallmark of HD pathology. The present study was directed at optimizing the potency of SIRT2 inhibitors containing the neuroprotective sulfobenzoic acid scaffold and improving their pharmacology. To achieve that goal, 176 analogues were designed, synthesized, and tested in deacetylation assays against the activities of major human sirtuins SIRT1-3. This screen yielded 15 compounds with enhanced potency for SIRT2 inhibition and 11 compounds having SIRT2 inhibition equal to reference compound AK-1. The newly synthesized compounds also demonstrated higher SIRT2 selectivity over SIRT1 and SIRT3. These candidates were subjected to a dose-response bioactivity assay, measuring an increase in α-tubulin K40 acetylation in two neuronal cell lines, which yielded five compounds bioactive in both cell lines and eight compounds bioactive in at least one of the cell lines tested. These bioactive compounds were subsequently tested in a tertiary polyglutamine aggregation assay, which identified five inhibitors. ADME properties of the bioactive SIRT2 inhibitors were assessed, which revealed a significant improvement of the pharmacological properties of the new entities, reaching closer to the goal of a clinically-viable candidate.


Subject(s)
Benzamides/pharmacology , Neuroprotective Agents/pharmacology , Sirtuin 2/antagonists & inhibitors , Acetylation , Benzamides/chemistry , Magnetic Resonance Spectroscopy , Mass Spectrometry , Neuroprotective Agents/chemistry , Sirtuin 2/metabolism , Structure-Activity Relationship
14.
Cell Rep ; 2(6): 1492-7, 2012 Dec 27.
Article in English | MEDLINE | ID: mdl-23200855

ABSTRACT

Inhibition of sirtuin 2 (SIRT2) deacetylase mediates protective effects in cell and invertebrate models of Parkinson's disease and Huntington's disease (HD). Here we report the in vivo efficacy of a brain-permeable SIRT2 inhibitor in two genetic mouse models of HD. Compound treatment resulted in improved motor function, extended survival, and reduced brain atrophy and is associated with marked reduction of aggregated mutant huntingtin, a hallmark of HD pathology. Our results provide preclinical validation of SIRT2 inhibition as a potential therapeutic target for HD and support the further development of SIRT2 inhibitors for testing in humans.


Subject(s)
Histone Deacetylase Inhibitors/pharmacology , Huntington Disease/drug therapy , Neuroprotective Agents/pharmacology , Sirtuin 2/antagonists & inhibitors , Animals , Disease Models, Animal , Drug Evaluation, Preclinical , Female , Huntington Disease/enzymology , Huntington Disease/genetics , Male , Mice , Mice, Mutant Strains , Sirtuin 2/genetics , Sirtuin 2/metabolism
15.
Bioorg Med Chem Lett ; 22(8): 2789-93, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22446090

ABSTRACT

Inhibition of sirtuin 2 (SIRT2) is known to be protective against the toxicity of disease proteins in Parkinson's and Huntington's models of neurodegeneration. Previously, we developed SIRT2 inhibitors based on the 3-(N-arylsulfamoyl)benzamide scaffold, including3-(N-(4-bromophenyl)sulfamoyl)-N-(4-bromophenyl)benzamide(C2-8, 1a), which demonstrated neuroprotective effects in a Huntington's mouse model, but had low potency of SIRT2 inhibition. Here we report that N-methylation of 1a greatly increases its potency and results in excellent selectivity for SIRT2 over SIRT1 and SIRT3 isoforms. Structure-activity relationships observed for 1a analogs and docking simulation data suggest that the para-substituted amido moiety of these compounds could occupy two potential hydrophobic binding pockets in SIRT2. These results provide a direction for the design of potent drug-like SIRT2 inhibitors.


Subject(s)
Benzamides/chemistry , Benzamides/pharmacology , Sirtuin 2/antagonists & inhibitors , Acylation , Animals , Binding Sites , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Humans , Hydrophobic and Hydrophilic Interactions , Mice , Molecular Conformation , Molecular Structure , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/pharmacology , Protein Isoforms , Sirtuin 1/chemistry , Sirtuin 3/chemistry , Structure-Activity Relationship , Sulfones/chemistry , Sulfones/pharmacology
16.
Hum Mol Genet ; 20(20): 3986-96, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21791548

ABSTRACT

Sirtuin 2 (SIRT2) is one of seven known mammalian protein deacetylases homologous to the yeast master lifespan regulator Sir2. In recent years, the sirtuin protein deacetylases have emerged as candidate therapeutic targets for many human diseases, including metabolic and age-dependent neurological disorders. In non-neuronal cells, SIRT2 has been shown to function as a tubulin deacetylase and a key regulator of cell division and differentiation. However, the distribution and function of the SIRT2 microtubule (MT) deacetylase in differentiated, postmitotic neurons remain largely unknown. Here, we show abundant and preferential expression of specific isoforms of SIRT2 in the mammalian central nervous system and find that a previously uncharacterized form, SIRT2.3, exhibits age-dependent accumulation in the mouse brain and spinal cord. Further, our studies reveal that focal areas of endogenous SIRT2 expression correlate with reduced α-tubulin acetylation in primary mouse cortical neurons and suggest that the brain-enriched species of SIRT2 may function as the predominant MT deacetylases in mature neurons. Recent reports have demonstrated an association between impaired tubulin acetyltransferase activity and neurodegenerative disease; viewed in this light, our results showing age-dependent accumulation of the SIRT2 neuronal MT deacetylase in wild-type mice suggest a functional link between tubulin acetylation patterns and the aging brain.


Subject(s)
Aging/metabolism , Central Nervous System/metabolism , Microtubules/metabolism , Neurons/metabolism , Sirtuin 2/metabolism , Animals , Cell Line, Tumor , Female , Gene Expression Regulation, Developmental , Gene Order , Humans , Male , Mice , Mice, Inbred C57BL , Microtubules/genetics , Protein Isoforms/metabolism , Sirtuin 2/genetics
17.
PLoS Curr ; 22010 Sep 02.
Article in English | MEDLINE | ID: mdl-20877454

ABSTRACT

The family of histone deacetylases (HDACs) has recently emerged as important drug targets for treatment of slow progressive neurodegenerative disorders, including Huntington's disease (HD). Broad pharmaceutical inhibition of HDACs has shown neuroprotective effects in various HD models. Here we examined the susceptibility of HDAC targets for drug treatment in affected brain areas during HD progression. We observed increased HDAC1 and decreased HDAC4, 5 and 6 levels, correlating with disease progression, in cortices and striata of HD R6/2 mice. However, there were no significant changes in HDAC protein levels, assessed in an age-dependent manner, in HD knock-in CAG140 mice and we did not observe significant changes in HDAC1 levels in human HD brains. We further assessed acetylation levels of α-tubulin, as a biomarker of HDAC6 activity, and found it unchanged in cortices from R6/2, knock-in, and human subjects at all disease stages. Inhibition of deacetylase activities was identical in cortical extracts from R6/2 and wild-type mice treated with a class II-selective HDAC inhibitor. Lastly, treatment with class I- and II-selective HDAC inhibitors showed similar responses in HD and wild-type rat striatal cells. In conclusion, our results show that class I and class II HDAC targets are present and accessible for chronic drug treatment during HD progression and provide impetus for therapeutic development of brain-permeable class- or isoform-selective inhibitors.

18.
Bone ; 44(2): 190-8, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19007918

ABSTRACT

Osteoclasts degrade bone matrix by demineralization followed by degradation of type I collagen through secretion of the cysteine protease, cathepsin K. Current imaging modalities are insufficient for sensitive observation of osteoclast activity, and in vivo live imaging of osteoclast resorption of bone has yet to be demonstrated. Here, we describe a near-infrared fluorescence reporter probe whose activation by cathepsin K is shown in live osteoclast cells and in mouse models of development and osteoclast upregulation. Cathepsin K probe activity was monitored in live osteoclast cultures and correlates with cathepsin K gene expression. In ovariectomized mice, cathepsin K probe upregulation precedes detection of bone loss by micro-computed tomography. These results are the first to demonstrate non-invasive visualization of bone degrading enzymes in models of accelerated bone loss, and may provide a means for early diagnosis of upregulated resorption and rapid feedback on efficacy of treatment protocols prior to significant loss of bone in the patient.


Subject(s)
Cathepsins/metabolism , Molecular Probe Techniques , Osteoclasts/enzymology , Animals , Animals, Newborn , Bone Development , Bone Resorption/enzymology , Calcification, Physiologic , Cathepsin K , Cathepsins/genetics , Cell Survival , Cells, Cultured , Cryoultramicrotomy , Female , Femur/pathology , Fluorescence , Humans , Mice , Mice, Inbred BALB C , Molecular Probes/metabolism , Osteoclasts/cytology , Ovariectomy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Up-Regulation
19.
Circulation ; 115(17): 2292-8, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17420353

ABSTRACT

BACKGROUND: Cathepsin K (CatK), a potent elastinolytic and collagenolytic cysteine protease, likely participates in the evolution and destabilization of atherosclerotic plaques. To assess better the biology of CatK activity in vivo, we developed a novel near-infrared fluorescence (NIRF) probe for imaging of CatK and evaluated it in mouse and human atherosclerosis. METHODS AND RESULTS: The NIRF imaging agent consists of the CatK peptide substrate GHPGGPQGKC-NH2 linked to an activatable fluorogenic polymer. In vitro, CatK produced a 2- to 14-fold activation of the agent over other cysteine and matrix metalloproteinases (P<0.0001), as well as a >8-fold activation over a control imaging agent (P<0.001). Optical imaging of atheroma revealed >100% NIRF signal increases in apolipoprotein E-/- mice in vivo (n=13; P<0.05, CatK imaging agent versus control agent) and in human carotid endarterectomy specimens ex vivo (n=14; P<0.05). Fluorescence microscopy of plaque sections demonstrated that enzymatically active CatK (positive NIRF signal) localized primarily in the vicinity of CatK-positive macrophages. Augmented NIRF signal (reflecting CatK activity) colocalized with disrupted elastin fibers within the media underlying plaques. CONCLUSIONS: Use of this novel protease-activatable NIRF agent for optical imaging in vivo demonstrated preferential localization of enzymatically active CatK to macrophages, consistent with their known greater elastinolytic capabilities compared with smooth muscle cells. Augmented CatK proteolysis in atheromata further links CatK to vascular remodeling and plaque vulnerability.


Subject(s)
Carotid Arteries/enzymology , Carotid Artery Diseases/metabolism , Cathepsins/metabolism , Microscopy, Fluorescence/methods , Spectroscopy, Near-Infrared/methods , Animals , Apolipoproteins E/genetics , Carotid Arteries/pathology , Carotid Artery Diseases/pathology , Cathepsin K , Elastin/metabolism , Enzyme Activation , Fluorescein-5-isothiocyanate , Fluorescent Dyes , Humans , Immunohistochemistry , Macrophages/enzymology , Macrophages/pathology , Mice , Mice, Mutant Strains , Peptide Hydrolases
20.
Mol Cancer Ther ; 5(8): 1944-9, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16928814

ABSTRACT

A potent cytotoxic peptide (r7-kla) was synthesized by incorporating a mitochondrial membrane disrupting peptide, kla (klaklakklaklak), with a cell-penetrating domain, r7 (rrrrrrr). The IC(50) of r7-kla (3.54 +/- 0.11 micromol/L) was more than two orders of magnitude lower than that of kla. r7-kla induced cell death in both in vitro and in vivo environments, and showed rapid kinetics. Within minutes, the morphologic changes in cells and mitochondrial leakage were apparent by microscopy and was consistent with rapid apoptosis. Our results suggested that r7-kla is an apoptosis inducer and can be potentially used as an antitumor agent, especially when combined with the appropriate systemic delivery systems.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Mitochondria/drug effects , Peptides/pharmacology , Recombinant Fusion Proteins/pharmacology , Animals , Cells, Cultured , Drug Screening Assays, Antitumor/methods , Female , Humans , Inhibitory Concentration 50 , Kinetics , Mice , Mice, Nude , Toxicity Tests , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...