Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Proc Natl Acad Sci U S A ; 120(16): e2217864120, 2023 04 18.
Article in English | MEDLINE | ID: mdl-37043533

ABSTRACT

Aberrant activity of cyclin-dependent kinase (Cdk5) has been implicated in various neurodegenerative diseases. This deleterious effect is mediated by pathological cleavage of the Cdk5 activator p35 into the truncated product p25, leading to prolonged Cdk5 activation and altered substrate specificity. Elevated p25 levels have been reported in humans and rodents with neurodegeneration, and the benefit of genetically blocking p25 production has been demonstrated previously in rodent and human neurodegenerative models. Here, we report a 12-amino-acid-long peptide fragment derived from Cdk5 (Cdk5i) that is considerably smaller than existing peptide inhibitors of Cdk5 (P5 and CIP) but shows high binding affinity toward the Cdk5/p25 complex, disrupts the interaction of Cdk5 with p25, and lowers Cdk5/p25 kinase activity. When tagged with a fluorophore (FITC) and the cell-penetrating transactivator of transcription (TAT) sequence, the Cdk5i-FT peptide exhibits cell- and brain-penetrant properties and confers protection against neurodegenerative phenotypes associated with Cdk5 hyperactivity in cell and mouse models of neurodegeneration, highlighting Cdk5i's therapeutic potential.


Subject(s)
Cyclin-Dependent Kinase 5 , Peptides , Mice , Animals , Humans , Cyclin-Dependent Kinase 5/metabolism , Phosphorylation , Peptides/metabolism , Peptide Fragments/metabolism , Phenotype
2.
J Neurochem ; 158(5): 1058-1073, 2021 09.
Article in English | MEDLINE | ID: mdl-34077555

ABSTRACT

Activity-regulated cytoskeleton-associated protein (Arc) is an immediate early gene product that support neuroplastic changes important for cognitive function and memory formation. As a protein with homology to the retroviral Gag protein, a particular characteristic of Arc is its capacity to self-assemble into virus-like capsids that can package mRNAs and transfer those transcripts to other cells. Although a lot has been uncovered about the contributions of Arc to neuron biology and behavior, very little is known about how different functions of Arc are coordinately regulated both temporally and spatially in neurons. The answer to this question we hypothesized must involve the occurrence of different protein post-translational modifications acting to confer specificity. In this study, we used mass spectrometry and sequence prediction strategies to map novel Arc phosphorylation sites. Our approach led us to recognize serine 67 (S67) and threonine 278 (T278) as residues that can be modified by TNIK, which is a kinase abundantly expressed in neurons that shares many functional overlaps with Arc and has, along with its interacting proteins such as the NMDA receptor, and been implicated as a risk factor for psychiatric disorders. Furthermore, characterization of each residue using site-directed mutagenesis to create S67 and T278 mutant variants revealed that TNIK action at those amino acids can strongly influence Arc's subcellular distribution and self-assembly as capsids. Together, our findings reveal an unsuspected connection between Arc and TNIK. Better understanding of the interplay between these two proteins in neuronal cells could lead to new insights about apparition and progression of psychiatric disorders. Cover Image for this issue: https://doi.org/10.1111/jnc.15077.


Subject(s)
Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Amino Acid Sequence , Animals , Cell Line, Tumor , Mice , Neurons/metabolism , Phosphorylation/physiology
3.
Bioorg Med Chem Lett ; 26(4): 1265-1271, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26804233

ABSTRACT

Targeting chromatin-mediated epigenetic regulation has emerged as a potential avenue for developing novel therapeutics for a wide range of central nervous system disorders, including cognitive disorders and depression. Histone deacetylase (HDAC) inhibitors have been pursued as cognitive enhancers that impact the regulation of gene expression and other mechanisms integral to neuroplasticity. Through systematic modification of the structure of crebinostat, a previously discovered cognitive enhancer that affects genes critical to memory and enhances synaptogenesis, combined with biochemical and neuronal cell-based screening, we identified a novel hydroxamate-based HDAC inhibitor, here named neurinostat, with increased potency compared to crebinostat in inducing neuronal histone acetylation. In addition, neurinostat was found to have a pharmacokinetic profile in mouse brain modestly improved over that of crebinostat. This discovery of neurinostat and demonstration of its effects on neuronal HDACs adds to the available pharmacological toolkit for dissecting the molecular and cellular mechanisms of neuroepigenetic regulation in health and disease.


Subject(s)
Biphenyl Compounds/chemistry , Brain/metabolism , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylases/metabolism , Hydrazines/chemistry , Acetylation , Animals , Biphenyl Compounds/chemical synthesis , Biphenyl Compounds/pharmacokinetics , Cells, Cultured , Half-Life , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/pharmacokinetics , Histone Deacetylases/chemistry , Histones/metabolism , Hydrazines/chemical synthesis , Hydrazines/pharmacokinetics , Inhibitory Concentration 50 , Mice , Neurons/cytology , Neurons/metabolism , Protein Binding , Structure-Activity Relationship
4.
Angew Chem Int Ed Engl ; 55(33): 9601-5, 2016 08 08.
Article in English | MEDLINE | ID: mdl-27355874

ABSTRACT

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.


Subject(s)
Brain/drug effects , Brain/diagnostic imaging , Neuroimaging , Oxazoles/pharmacology , Positron-Emission Tomography , Protein Kinase Inhibitors/pharmacology , Triazoles/pharmacology , tau Proteins/antagonists & inhibitors , Brain/metabolism , Crystallography, X-Ray , Dose-Response Relationship, Drug , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Humans , Models, Molecular , Molecular Structure , Oxazoles/chemical synthesis , Oxazoles/chemistry , Phosphorylation/drug effects , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Triazoles/chemical synthesis , Triazoles/chemistry , tau Proteins/metabolism
5.
Sci Rep ; 14(1): 9064, 2024 04 20.
Article in English | MEDLINE | ID: mdl-38643236

ABSTRACT

Frontotemporal dementia (FTD) is a debilitating neurodegenerative disorder with currently no disease-modifying treatment options available. Mutations in GRN are one of the most common genetic causes of FTD, near ubiquitously resulting in progranulin (PGRN) haploinsufficiency. Small molecules that can restore PGRN protein to healthy levels in individuals bearing a heterozygous GRN mutation may thus have therapeutic value. Here, we show that epigenetic modulation through bromodomain and extra-terminal domain (BET) inhibitors (BETi) potently enhance PGRN protein levels, both intracellularly and secreted forms, in human central nervous system (CNS)-relevant cell types, including in microglia-like cells. In terms of potential for disease modification, we show BETi treatment effectively restores PGRN levels in neural cells with a GRN mutation known to cause PGRN haploinsufficiency and FTD. We demonstrate that BETi can rapidly and durably enhance PGRN in neural progenitor cells (NPCs) in a manner dependent upon BET protein expression, suggesting a gain-of-function mechanism. We further describe a CNS-optimized BETi chemotype that potently engages endogenous BRD4 and enhances PGRN expression in neuronal cells. Our results reveal a new epigenetic target for treating PGRN-deficient forms of FTD and provide mechanistic insight to aid in translating this discovery into therapeutics.


Subject(s)
Frontotemporal Dementia , Humans , Progranulins/metabolism , Frontotemporal Dementia/drug therapy , Frontotemporal Dementia/genetics , Frontotemporal Dementia/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Mutation , Epigenesis, Genetic , Bromodomain Containing Proteins , Cell Cycle Proteins/metabolism
6.
Bioorg Med Chem Lett ; 22(5): 2015-9, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22335895

ABSTRACT

Haspin is a serine/threonine kinase that phosphorylates Thr-3 of histone H3 in mitosis that has emerged as a possible cancer therapeutic target. High throughput screening of approximately 140,000 compounds identified the beta-carbolines harmine and harmol as moderately potent haspin kinase inhibitors. Based on information obtained from a structure-activity relationship study previously conducted for an acridine series of haspin inhibitors in conjunction with in silico docking using a recently disclosed crystal structure of the kinase, harmine analogs were designed that resulted in significantly increased haspin kinase inhibitory potency. The harmine derivatives also demonstrated less activity towards DYRK2 compared to the acridine series. In vitro mouse liver microsome stability and kinase profiling of a representative member of the harmine series (42, LDN-211898) are also presented.


Subject(s)
Carbolines/chemistry , Carbolines/pharmacology , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Carbolines/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Microsomes, Liver/metabolism , Models, Molecular , Protein Kinase Inhibitors/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Structure-Activity Relationship , Dyrk Kinases
7.
Proc Natl Acad Sci U S A ; 106(48): 20198-203, 2009 12 01.
Article in English | MEDLINE | ID: mdl-19918057

ABSTRACT

The protein kinase haspin/Gsg2 plays an important role in mitosis, where it specifically phosphorylates Thr-3 in histone H3 (H3T3). Its protein sequence is only weakly homologous to other protein kinases and lacks the highly conserved motifs normally required for kinase activity. Here we report structures of human haspin in complex with ATP and the inhibitor iodotubercidin. These structures reveal a constitutively active kinase conformation, stabilized by haspin-specific inserts. Haspin also has a highly atypical activation segment well adapted for specific recognition of the basic histone tail. Despite the lack of a DFG motif, ATP binding to haspin is similar to that in classical kinases; however, the ATP gamma-phosphate forms hydrogen bonds with the conserved catalytic loop residues Asp-649 and His-651, and a His651Ala haspin mutant is inactive, suggesting a direct role for the catalytic loop in ATP recognition. Enzyme kinetic data show that haspin phosphorylates substrate peptides through a rapid equilibrium random mechanism. A detailed analysis of histone modifications in the neighborhood of H3T3 reveals that increasing methylation at Lys-4 (H3K4) strongly decreases substrate recognition, suggesting a key role of H3K4 methylation in the regulation of haspin activity.


Subject(s)
Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Models, Molecular , Protein Conformation , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Adenosine Triphosphate/metabolism , Histones/metabolism , Humans , Phosphorylation , Tubercidin/metabolism
8.
ACS Chem Neurosci ; 12(2): 271-284, 2021 01 20.
Article in English | MEDLINE | ID: mdl-33417763

ABSTRACT

Genomic instability caused by a deficiency in the DNA damage response and repair has been linked to age-related cognitive decline and neurodegenerative diseases. Preventing genomic instability that ultimately leads to neuronal death may provide a broadly effective strategy to protect against multiple potential genotoxic stressors. Recently, the zinc-dependent class I histone deacetylase (HDAC1) has been identified as a critical factor for protecting neurons from deleterious effects of DNA damage in Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), and frontotemporal dementia (FTD). Translating these observations to a novel neuroprotective therapy for AD, ALS, and FTD may be advanced by the identification of small molecules capable of increasing the deacetylase activity of HDAC1 selectively over other structurally similar HDACs. Here, we demonstrate that exifone, a drug previously shown to be effective in treating cognitive deficits associated with AD and Parkinson's disease, the molecular mechanism of which has remained poorly understood, potently activates the deacetylase activity of HDAC1. We show that exifone acts as a mixed, nonessential activator of HDAC1 that is capable of binding to both free and substrate-bound enzyme, resulting in an increased relative maximal rate of HDAC1-catalyzed deacetylation. Exifone can directly bind to HDAC1 based upon biolayer interferometry assays with kinetic and selectivity profiling, suggesting that HDAC1 is preferentially targeted compared to other class I HDACs and the kinase CDK5, which have also been implicated in neurodegeneration. Consistent with a mechanism of deacetylase activation intracellularly, the treatment of human induced pluripotent stem cell (iPSC)-derived neuronal cells resulted in globally decreased histone acetylation. Moreover, exifone treatment was neuroprotective in a tauopathy patient iPSC-derived neuronal model subject to oxidative stress. Taken together, these findings reveal exifone as a potent activator of HDAC1-mediated deacetylation, thereby offering a lead for novel therapeutic development aiming to protect genomic integrity in the context of neurodegeneration and aging.


Subject(s)
Histone Deacetylases , Induced Pluripotent Stem Cells , Benzophenones , Histone Deacetylase 1 , Humans , Neurons
9.
J Med Chem ; 64(19): 14745-14756, 2021 10 14.
Article in English | MEDLINE | ID: mdl-34549949

ABSTRACT

To better understand the role of bromodomain and extra-terminal domain (BET) proteins in epigenetic mechanisms, we developed a series of thienodiazepine-based derivatives and identified two compounds, 3a and 6a, as potent BET inhibitors. Further in vivo pharmacokinetic studies and analysis of in vitro metabolic stability of 6a revealed excellent brain penetration and reasonable metabolic stability. Compounds 3a and 6a were radiolabeled with fluorine-18 in two steps and utilized in positron emission tomography (PET) imaging studies in mice. Preliminary PET imaging results demonstrated that [18F]3a and [18F]6a have good brain uptake (with maximum SUV = 1.7 and 2, respectively) and binding specificity in mice brains. These results show that [18F]6a is a potential PET radiotracer that could be applied to imaging BET proteins in the brain. Further optimization and improvement of the metabolic stability of [18F]6a are still needed in order to create optimal PET imaging probes of BET family members.


Subject(s)
Azepines/chemistry , Drug Design , Molecular Probes/chemistry , Positron-Emission Tomography/methods , Protein Domains , Animals , Azepines/pharmacokinetics , Mice , Molecular Docking Simulation , Molecular Probes/pharmacokinetics , Transcription Factors/metabolism
10.
Sci Rep ; 11(1): 17029, 2021 08 23.
Article in English | MEDLINE | ID: mdl-34426604

ABSTRACT

Mutations in MAPT (microtubule-associated protein tau) cause frontotemporal dementia (FTD). MAPT mutations are associated with abnormal tau phosphorylation levels and accumulation of misfolded tau protein that can propagate between neurons ultimately leading to cell death (tauopathy). Recently, a p.A152T tau variant was identified as a risk factor for FTD, Alzheimer's disease, and synucleinopathies. Here we used induced pluripotent stem cells (iPSC) from a patient carrying this p.A152T variant to create a robust, functional cellular assay system for probing pathophysiological tau accumulation and phosphorylation. Using stably transduced iPSC-derived neural progenitor cells engineered to enable inducible expression of the pro-neural transcription factor Neurogenin 2 (Ngn2), we generated disease-relevant, cortical-like glutamatergic neurons in a scalable, high-throughput screening compatible format. Utilizing automated confocal microscopy, and an advanced image-processing pipeline optimized for analysis of morphologically complex human neuronal cultures, we report quantitative, subcellular localization-specific effects of multiple kinase inhibitors on tau, including ones under clinical investigation not previously reported to affect tau phosphorylation. These results demonstrate the potential for using patient iPSC-derived ex vivo models of tauopathy as genetically accurate, disease-relevant systems to probe tau biochemistry and support the discovery of novel therapeutics for tauopathies.


Subject(s)
Glutamates/metabolism , Image Processing, Computer-Assisted , Induced Pluripotent Stem Cells/metabolism , Models, Biological , Neurons/pathology , Proteomics , Tauopathies/pathology , tau Proteins/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Biomarkers/metabolism , Cell Line , Humans , Induced Pluripotent Stem Cells/drug effects , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Phosphorylation/drug effects , Protein Kinases/metabolism , Pyridines/chemistry , Pyridines/pharmacology , Pyrimidines/chemistry , Pyrimidines/pharmacology , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology
11.
Bioorg Med Chem Lett ; 20(12): 3491-4, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20836251

ABSTRACT

Haspin is a serine/threonine kinase required for completion of normal mitosis that is highly expressed during cell proliferation, including in a number of neoplasms. Consequently, it has emerged as a potential therapeutic target in oncology. A high throughput screen of approximately 140,000 compounds identified an acridine analog as a potent haspin kinase inhibitor. Profiling against a panel of 270 kinases revealed that the compound also exhibited potent inhibitory activity for DYRK2, another serine/threonine kinase. An optimization study of the acridine series revealed that the structure-activity relationship (SAR) of the acridine series for haspin and DYRK2 inhibition had many similarities. However, several structural differences were noted that allowed generation of a potent haspin kinase inhibitor (33, IC50 <60 nM) with 180-fold selectivity over DYRK2. In addition, a moderately potent DYRK2 inhibitor (41, IC50 <400 nM) with a 5.4-fold selectivity over haspin was also identified.


Subject(s)
Acridines/pharmacology , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Acridines/chemical synthesis , Humans , Inhibitory Concentration 50 , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship , Dyrk Kinases
12.
BMC Res Notes ; 13(1): 254, 2020 May 24.
Article in English | MEDLINE | ID: mdl-32448288

ABSTRACT

OBJECTIVES: UHRF1 is a multi-domain protein that recognizes both histone and DNA modification marks on chromatin. UHRF1 is involved in various cellular processes that lead to tumorigenesis and thus attracted considerable attention as a potential anti-cancer drug target. The SRA domain is a unique to the UHRF family. SRA domain recognizes 5-methylcytosine in hemimethylated DNA and necessary for maintenance DNA methylation mediated by DNMT1. Small molecules capable of interacting with the SRA domain may reduce aberrant methylation levels by preventing the interaction of 5-methylcytosine with the SRA domain and thereby blocking substrate access to the catalytic center of DNMT1. The data were collected to identify and predict an initial set of small molecules that are expected to bind to the SRA domain. DATA DESCRIPTION: Nearly 2.4 million molecules from various chemical libraries were screened with the SRA domain of UHRF1 using Schrodinger's Small Molecule Drug Discovery Suite. The data is available in the form of a methodology presentation, MS Excel files listing the top hits, and Maestro pose viewer files that provide visualization of how the identified ligands interact with the SRA domain.


Subject(s)
CCAAT-Enhancer-Binding Proteins/antagonists & inhibitors , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , Small Molecule Libraries/chemistry , Ubiquitin-Protein Ligases/antagonists & inhibitors , 5-Methylcytosine/chemistry , CCAAT-Enhancer-Binding Proteins/chemistry , Catalytic Domain/drug effects , DNA (Cytosine-5-)-Methyltransferase 1/chemistry , DNA Methylation/drug effects , Humans , Protein Binding , Protein Domains , Ubiquitin-Protein Ligases/chemistry
13.
Nucl Med Biol ; 84-85: 96-101, 2020.
Article in English | MEDLINE | ID: mdl-32320910

ABSTRACT

INTRODUCTION: Bromodomain and extra-terminal domain (BET) family proteins play a vital role in the epigenetic regulation process by interacting with acetylated lysine (Ac-K) residues in histones. BET inhibitors have become promising candidates to treat various diseases through the inhibition of the interaction between BET bromodomains and Ac-K of histone tails. With a molecular imaging probe, noninvasive imaging such as positron emission tomography (PET) can visualize the distribution and roles of BET family proteins in vivo and enlighten our understanding of BET protein function in both healthy and diseased tissue. METHODS: We radiolabeled the potent BET inhibitor INCB054329 by N-methylation to make [11C]PB003 as a BET PET radiotracer. The bioactivity evaluation of unlabeled PB003 in vitro was performed to confirm its binding affinity for BRDs, then the PET/CT imaging in rodents was performed to evaluate the bioactivity of [11C]PB003 in vivo. RESULTS: In our in vitro evaluation, PB003 showed a high BET binding affinity for BRDs (Kd = 2 nM, 1.2 nM, and 1.2 nM for BRD2, BRD3, and BRD4, respectively). In vivo PET/CT imaging demonstrated that [11C]PB003 has favorable uptake with appropriate kinetics and distributions in main peripheral organs. Besides, the blockade of [11C]PB003 binding was found in our blocking study which indicated the specificity of [11C]PB003. However, the BBB penetration and brain uptake of [11C]PB003 was limited, with only a maximum 0.2% injected dose/g at ~2 min post-injection. CONCLUSION: The imaging results in rodents in vivo demonstrate that [11C]PB003 binds to BET with high selectivity and specificity and has favorable uptake in peripheral organs. However, the low brain uptake of [11C]PB003 limits the visualization of brain regions indicating the efforts are still needed to discover the new BET imaging probes for brain visualization.


Subject(s)
Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Positron-Emission Tomography/methods , Brain/diagnostic imaging , Brain/metabolism , Isotope Labeling , Kinetics , Methylation , Protein Domains , Radioactive Tracers , Radiochemistry
14.
ACS Chem Neurosci ; 11(24): 4128-4138, 2020 12 16.
Article in English | MEDLINE | ID: mdl-33253521

ABSTRACT

Glycogen synthase kinase 3-beta (GSK3ß) is a critical regulator of several cellular pathways involved in neurodevelopment and neuroplasticity and as such is a potential focus for the discovery of new neurotherapeutics toward the treatment of neuropsychiatric and neurodegenerative diseases. The majority of efforts to develop inhibitors of GSK3ß have been focused on developing small molecule inhibitors that compete with adenosine triphosphate (ATP) through direct interaction with the ATP binding site. This strategy has presented selectivity challenges due to the evolutionary conservation of this domain within the kinome. The disrupted in schizophrenia 1 (DISC1) protein has previously been shown to bind and inhibit GSK3ß activity. Here, we report the characterization of a 44-mer peptide derived from human DISC1 (hDISCtide) that is sufficient to both bind and inhibit GSK3ß in a noncompetitive mode distinct from classical ATP competitive inhibitors. Based on multiple independent biochemical and biophysical assays, we propose that hDISCtide interacts at two distinct regions of GSK3ß: an inhibitory region that partially overlaps with the binding site of FRATide, a well-known GSK3ß binding peptide, and a specific binding region that is unique to hDISCtide. Taken together, our findings present a novel avenue for developing a peptide-based selective inhibitor of GSK3ß.


Subject(s)
Glycogen Synthase Kinase 3 beta , Nerve Tissue Proteins , Humans , Nerve Tissue Proteins/metabolism , Peptides/pharmacology , Phosphorylation
15.
Nat Commun ; 11(1): 2484, 2020 05 18.
Article in English | MEDLINE | ID: mdl-32424276

ABSTRACT

DNA damage contributes to brain aging and neurodegenerative diseases. However, the factors stimulating DNA repair to stave off functional decline remain obscure. We show that HDAC1 modulates OGG1-initated 8-oxoguanine (8-oxoG) repair in the brain. HDAC1-deficient mice display age-associated DNA damage accumulation and cognitive impairment. HDAC1 stimulates OGG1, a DNA glycosylase known to remove 8-oxoG lesions that are associated with transcriptional repression. HDAC1 deficiency causes impaired OGG1 activity, 8-oxoG accumulation at the promoters of genes critical for brain function, and transcriptional repression. Moreover, we observe elevated 8-oxoG along with reduced HDAC1 activity and downregulation of a similar gene set in the 5XFAD mouse model of Alzheimer's disease. Notably, pharmacological activation of HDAC1 alleviates the deleterious effects of 8-oxoG in aged wild-type and 5XFAD mice. Our work uncovers important roles for HDAC1 in 8-oxoG repair and highlights the therapeutic potential of HDAC1 activation to counter functional decline in brain aging and neurodegeneration.


Subject(s)
Aging/pathology , Alzheimer Disease/pathology , Brain/pathology , DNA Damage , DNA Glycosylases/metabolism , Histone Deacetylase 1/metabolism , Oxidative Stress , Acetylation , Aging/genetics , Alzheimer Disease/complications , Alzheimer Disease/physiopathology , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Base Sequence , Benzophenones/pharmacology , Cognition/drug effects , Cognition Disorders/complications , Cognition Disorders/pathology , Down-Regulation/drug effects , Down-Regulation/genetics , Gene Ontology , Guanine/analogs & derivatives , Guanine/metabolism , Memory/drug effects , Mice, Inbred C57BL , Mice, Knockout , Neurons/drug effects , Neurons/metabolism , Oxidative Stress/drug effects , Promoter Regions, Genetic/genetics
16.
Transl Psychiatry ; 10(1): 76, 2020 02 24.
Article in English | MEDLINE | ID: mdl-32094324

ABSTRACT

The effective treatment of bipolar disorder (BD) represents a significant unmet medical need. Although lithium remains a mainstay of treatment for BD, limited knowledge regarding how it modulates affective behavior has proven an obstacle to discovering more effective mood stabilizers with fewer adverse side effects. One potential mechanism of action of lithium is through inhibition of the serine/threonine protein kinase GSK3ß, however, relevant substrates whose change in phosphorylation may mediate downstream changes in neuroplasticity remain poorly understood. Here, we used human induced pluripotent stem cell (hiPSC)-derived neuronal cells and stable isotope labeling by amino acids in cell culture (SILAC) along with quantitative mass spectrometry to identify global changes in the phosphoproteome upon inhibition of GSK3α/ß with the highly selective, ATP-competitive inhibitor CHIR-99021. Comparison of phosphorylation changes to those induced by therapeutically relevant doses of lithium treatment led to the identification of collapsin response mediator protein 2 (CRMP2) as being highly sensitive to both treatments as well as an extended panel of structurally distinct GSK3α/ß inhibitors. On this basis, a high-content image-based assay in hiPSC-derived neurons was developed to screen diverse compounds, including FDA-approved drugs, for their ability to mimic lithium's suppression of CRMP2 phosphorylation without directly inhibiting GSK3ß kinase activity. Systemic administration of a subset of these CRMP2-phosphorylation suppressors were found to mimic lithium's attenuation of amphetamine-induced hyperlocomotion in mice. Taken together, these studies not only provide insights into the neural substrates regulated by lithium, but also provide novel human neuronal assays for supporting the development of mechanism-based therapeutics for BD and related neuropsychiatric disorders.


Subject(s)
Bipolar Disorder , Induced Pluripotent Stem Cells , Amphetamine/pharmacology , Animals , Bipolar Disorder/drug therapy , Humans , Lithium/pharmacology , Lithium Compounds/pharmacology , Mice , Phosphorylation
17.
Nucleic Acids Res ; 35(21): 7313-23, 2007.
Article in English | MEDLINE | ID: mdl-17962312

ABSTRACT

Methylation of lysine residues on histones participates in transcriptional gene regulation. Lysine 9 methylation of histone H3 is a transcriptional repression signal, mediated by a family of SET domain containing AdoMet-dependent enzymes. G9a methyltransferase is a euchromatic histone H3 lysine 9 methyltransferase. Here, G9a is shown to methylate other cellular proteins, apart from histone H3, including automethylation of K239 residue. Automethylation of G9a did not impair or activate the enzymatic activity in vitro. The automethylation motif of G9a flanking target K239 (ARKT) has similarity with histone H3 lysine 9 regions (ARKS), and is identical to amino acids residues in EuHMT (ARKT) and mAM (ARKT). Under steady-state kinetic assay conditions, full-length G9a methylates peptides representing ARKS/T motif of H3, G9a, mAM and EuHMT efficiently. Automethylation of G9a at ARKT motif creates a binding site for HP1 class of protein and mutation of lysine in the motif impairs this binding. In COS-7 cells GFP fusion of the wild-type G9a co-localized with HP1alpha and HP1gamma isoforms whereas the G9a mutant with K239A displayed poor co-localization. Thus, apart from transcriptional repression and regulatory roles of lysine methylation, the non-histone protein methylation may create binding sites for cellular protein-protein interactions.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , COS Cells , Cell Line , Chlorocebus aethiops , Chromobox Protein Homolog 5 , Histone-Lysine N-Methyltransferase/chemistry , Histone-Lysine N-Methyltransferase/genetics , Lysine/metabolism , Mass Spectrometry , Methylation , Mice , Molecular Sequence Data , Mutation , Protein Processing, Post-Translational , Substrate Specificity
18.
Chem Commun (Camb) ; 55(86): 12932-12935, 2019 Oct 24.
Article in English | MEDLINE | ID: mdl-31599282

ABSTRACT

Here, we report the development of novel PET radiotracer ([11C]CW22) of BET proteins. In vivo imaging results in rodents and nonhuman primates (NHP) demonstrate that [11C]CW22 has excellent brain uptake, good specificity, good selectivity, suitable metabolism, appropriate kinetics and distribution in the brain. Our studies demonstrated that [11C]CW22 exhibits ideal properties as a PET imaging probe of BET proteins for further validation.


Subject(s)
Brain/diagnostic imaging , Positron-Emission Tomography , Radiopharmaceuticals/chemistry , Animals , Blood-Testis Barrier/metabolism , Brain/metabolism , Carbon Radioisotopes/chemistry , Crystallography, X-Ray , Kinetics , Macaca , Mice , Molecular Conformation , Nerve Tissue Proteins/chemistry , Neurons , Protein Domains , Radiopharmaceuticals/metabolism , Receptors, Cell Surface/chemistry
19.
Elife ; 82019 03 25.
Article in English | MEDLINE | ID: mdl-30907729

ABSTRACT

Tauopathies are neurodegenerative diseases characterized by aberrant forms of tau protein accumulation leading to neuronal death in focal brain areas. Positron emission tomography (PET) tracers that bind to pathological tau are used in diagnosis, but there are no current therapies to eliminate these tau species. We employed targeted protein degradation technology to convert a tau PET-probe into a functional degrader of pathogenic tau. The hetero-bifunctional molecule QC-01-175 was designed to engage both tau and Cereblon (CRBN), a substrate-receptor for the E3-ubiquitin ligase CRL4CRBN, to trigger tau ubiquitination and proteasomal degradation. QC-01-175 effected clearance of tau in frontotemporal dementia (FTD) patient-derived neuronal cell models, with minimal effect on tau from neurons of healthy controls, indicating specificity for disease-relevant forms. QC-01-175 also rescued stress vulnerability in FTD neurons, phenocopying CRISPR-mediated MAPT-knockout. This work demonstrates that aberrant tau in FTD patient-derived neurons is amenable to targeted degradation, representing an important advance for therapeutics.


Subject(s)
Frontotemporal Dementia/drug therapy , Neurons/metabolism , Neuroprotective Agents/metabolism , Proteolysis , Tauopathies/drug therapy , tau Proteins/metabolism , Cell Line , Humans , Models, Theoretical , Neurons/drug effects , Neuroprotective Agents/chemical synthesis
20.
J Med Chem ; 62(21): 9600-9617, 2019 11 14.
Article in English | MEDLINE | ID: mdl-31535859

ABSTRACT

Using structure-guided design, several cell based assays, and microdosed positron emission tomography (PET) imaging, we identified a series of highly potent, selective, and brain-penetrant oxazole-4-carboxamide-based inhibitors of glycogen synthase kinase-3 (GSK-3). An isotopologue of our first-generation lead, [3H]PF-367, demonstrates selective and specific target engagement in vitro, irrespective of the activation state. We discovered substantial ubiquitous GSK-3-specific radioligand binding in Tg2576 Alzheimer's disease (AD), suggesting application for these compounds in AD diagnosis and identified [11C]OCM-44 as our lead GSK-3 radiotracer, with optimized brain uptake by PET imaging in nonhuman primates. GSK-3ß-isozyme selectivity was assessed to reveal OCM-51, the most potent (IC50 = 0.030 nM) and selective (>10-fold GSK-3ß/GSK-3α) GSK-3ß inhibitor known to date. Inhibition of CRMP2T514 and tau phosphorylation, as well as favorable therapeutic window against WNT/ß-catenin signaling activation, was observed in cells.


Subject(s)
Brain/metabolism , Drug Discovery , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Positron-Emission Tomography/methods , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Animals , Blood-Brain Barrier/metabolism , Brain/diagnostic imaging , Catalytic Domain , Glycogen Synthase Kinase 3 beta/chemistry , HEK293 Cells , Humans , Mice , Models, Molecular , Neuroimaging , Oxazoles/chemistry , Oxazoles/metabolism , Oxazoles/pharmacology , Protein Kinase Inhibitors/metabolism , Triazoles/chemistry , Triazoles/metabolism , Triazoles/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL