In Vivo Imaging of Histone Deacetylases (HDACs) in the Central Nervous System and Major Peripheral Organs
- Changning Wang
- ,
- Frederick A. Schroeder
- ,
- Hsiao-Ying Wey
- ,
- Ronald Borra
- ,
- Florence F. Wagner
- ,
- Surya Reis
- ,
- Sung Won Kim
- ,
- Edward B. Holson
- ,
- Stephen J. Haggarty
- , and
- Jacob M. Hooker
Abstract

Epigenetic enzymes are now targeted to treat the underlying gene expression dysregulation that contribute to disease pathogenesis. Histone deacetylases (HDACs) have shown broad potential in treatments against cancer and emerging data supports their targeting in the context of cardiovascular disease and central nervous system dysfunction. Development of a molecular agent for non-invasive imaging to elucidate the distribution and functional roles of HDACs in humans will accelerate medical research and drug discovery in this domain. Herein, we describe the synthesis and validation of an HDAC imaging agent, [11C]6. Our imaging results demonstrate that this probe has high specificity, good selectivity, and appropriate kinetics and distribution for imaging HDACs in the brain, heart, kidney, pancreas, and spleen. Our findings support the translational potential for [11C]6 for human epigenetic imaging.
Introduction
disease/process | selected evidence of HDAC 1, 2, 3, and 6 involvement | refs |
---|---|---|
neurodegenerative disorders | HDAC 1 and 3 protein complex integrity links deficient DNA repair with neurotoxicity | 1, 2 |
HDAC 1 expression is elevated in vulnerable brain regions of mouse disease models | 3 | |
HDAC 2 is elevated in Alzheimer’s patient brain and negatively impacts memory in animal models | 4 | |
HDAC 6 overexpression in patients with AD | 5 | |
learning and memory | HDAC 1 overexpression in the prefrontal cortex disrupts working memory | 6 |
HDAC 2 loss improves working memory and accelerates extinction learning | 7 | |
heart failure | HDAC 1 and 2 are critical mediators of autophagy and cardiac plasticity | 8 |
Class I HDAC enzymes alter cardiomyocyte hypertrophy via ERK kinase activity | 9 | |
asthma | HDAC 2 deficiency in cells blunts transcriptional response to steroids and anti-inflammatory drugs. | 10 |
cancer (general) | HDAC acetylation of p53 mediates tumor cell survival | 11 |
breast cancer | HDAC 6 mRNA expression may have potential both as a marker of endocrine responsiveness and as a prognostic indicator in breast cancer | 12 |
colon cancer | HDAC 3 turnover is induced in cells by isothiocyanate therapeutics | 13 |
HDAC 1 and NuRD complex contributes to epigenetic silencing of colorectal tumor suppressor genes | 14 | |
ovarian cancer | HDACs 1–3 overexpression confer cisplatin resistance in ovarian cancer cell lines | 15 |
HDAC 1 and 3 overexpression mediate proliferation and migration of ovarian cancer cells | 16 |
Figure 1

Figure 1. [11C]6: a translational PET imaging probe. We have developed a potent HDAC imaging agent, termed [11C]6, incorporating three key structural features to create a versatile and translational probe for visualizing HDAC expression in vivo. Intravenous injection of trace amounts of [11C]6 (nanogram scale) in baboon and imaging by PET-MR demonstrates quantifiable uptake in the brain and in diverse peripheral organs. This illustrates the potential for [11C]6 as a broadly applicable tool in evaluating HDAC density in humans.
Results
Design and Synthesis of PET Imaging Agents with Brain Permeability
Scheme 1

Scheme aReagents and conditions: (a) NaBH4, MeOH, overnight, rt, 75%; (b) formaldehyde, AcOH, NaBH4, MeOH, rt, overnight, 55%. (c) NH2OH (aq), 1M NaOH, MeOH/THF, 0 °C to rt, 4 h, 42% for 6, 40% for 5; RCY of [11C]6, 3–5% (non-decay corrected to trapped [11C]CH3I).
Physicochemical Properties of 6 and ex Vivo Binding
assay | 6 | SAHA |
---|---|---|
IC50 (nM) | ||
HDAC 1 | 0.3 | 4.0 |
HDAC 2 | 2.0 | 11 |
HDAC 3 | 0.6 | 3.0 |
HDAC 4 | 1970 | >30000 |
HDAC 5 | 352 | 8750 |
HDAC 6 | 4.1 | 2.0 |
HDAC 7 | >20000 | >30000 |
HDAC 8 | >15000 | 1020 |
HDAC 9 | >15000 | >30000 |
EC50 (nM) | ||
H3K9ac | 100 | 3400 |
H4K12ac | 100 | 1900 |
In vitro IC50 (nM) values using recombinant human enzymes for HDAC subtypes 1–9 and specific substrates demonstrate that 6 is a selective inhibitor of HDAC 1–3 (0.3–2.0 nM) with decreased potency to inhibit HDAC 6 (4.1 nM) or other subtypes (>352 nM). Comparatively, the hydroxamate HDAC inhibitor SAHA exhibited lower affinity for HDAC targets 1–3 (3.0–11 nM). Dose–response plots in cultured primary mouse neuronal cells measuring relative H3K9ac and H4K12ac levels revealed potent induction of histone acetylation (EC50) by 6 (100 nM) compared to SAHA (1900–3400 nM).
In Vivo PET-CT Imaging with [11C]6 in Rodents
High Brain Uptake and Reversible Binding
Dose-Dependent Blockade
In Vivo PET-MR Imaging with [11C]6 in Baboon
High Uptake in the Brain and Dose-Dependent Blockade
Figure 2

Figure 2. Kinetic modeling results with [11C]6 in baboon brain. (A) The total volume of distribution (VT) images from one representative animal show robust differences in radiotracer uptake at baseline and after blocking (0.5 mg/kg iv, 10 min pretreatment); (B) Two independent baseline-blocking studies were used to resolve quantitative VT data which show that pretreatment with unlabeled 6 (0.5 or 1.0 mg/kg) dose-dependently blocks tracer uptake in different baboon brain regions. WB: whole brain; CB: cerebellum; M1: primary motor cortex ; PU: putamen; TH: thalamus; V1: primary visual cortex ; CA: caudate; WM: white matter.
Arterial Plasma Analysis
Kinetic Modeling Shows High Specific Binding
High uptake and blockade in peripheral organs
Figure 3

Figure 3. [11C]6 PET-MR imaging. Axial views of summed PET images (40–80 min) superimposed with MR images from the same baboon following injection of radiotracer (4 mCi/baboon). Images illustrate tracer uptake in organs of interest at baseline and after pretreatment with unlabeled 6 (0.5 mg/kg). Robust blocking was observed in organs of interest including: A, heart; B, spleen; C, kidneys; and D, pancreas. Time–activity curves (baseline, blue; blocking, red) demonstrate a high specific binding of [11C]6 in these peripheral organs as the percent injected tracer dose per cm3 tissue is markedly reduced by blocking.
Discussion
Experimental Section
General Methods and Materials
Chemical Synthesis
(E)-Methyl 3-(4-((((3r,5r,7r)-adamantan-1-ylmethyl)amino)methyl)phenyl)acrylate (3)
(E)-3-(4-((((3r,5r,7r)-Adamantan-1-ylmethyl)amino)methyl)phenyl)-N-hydroxyacrylamide (4)
(E)-Methyl 3-(4-((((3r,5r,7r)-adamantan-1-ylmethyl) (methyl) amino) methyl) phenyl) acrylate (5)
(E)-3-(4-((((3r,5r,7r)-Adamantan-1-ylmethyl)(methyl)amino)methyl)phenyl)-N-hydroxyacrylamide (6)
HDAC Inhibition Assay
Mouse Primary Neuronal Histone Acetylation Assays
CNS Target Binding Assay
Radiosynthesis of [11C]6
Specific Activity of [11C]6
Log D Determination
Plasma Protein Binding Assay
Ex vivo Autoradiography
Rodent PET-CT Acquisition and Post-Processing
Rodent PET-CT Image Analysis
Baboon PET-MR Acquisition
Brain imaging
Body imaging
Baboon PET-MR Image Analysis

Plasma and Metabolite Analysis
Brain Metabolite Analysis
Supporting Information
The results of off-target binding, mPET imaging in rodents and SAHA pretreated study in NHP were shown in Table S1, Figure S1–S6. This material is available free of charge via the Internet at http://pubs.acs.org.
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
Acknowledgment
C.W. and H.Y.W. are supported by the Harvard/MGH Nuclear Medicine Training Program from the Department of Energy (DE-SC0008430). Research was supported by the National Institute of Drug Abuse (NIDA) of the National Institutes of Health under grant numbers R01DA030321 (J.M.H.; S.J.H) with additional support under grant R01DA028301 (S.J.H.). This research was carried out at the Athinoula A. Martinos Center for Biomedical Imaging at the Massachusetts General Hospital, using resources provided by the Center for Functional Neuroimaging Technologies, P41EB015896, a P41 Regional Resource supported by the National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health. This work also involved the use of instrumentation supported by the NIH Shared Instrumentation Grant Program and/or High-End Instrumentation Grant Program; specifically, grant nos: S10RR017208, S10RR026666, S10RR022976, S10RR019933, and S10RR029495. The authors are grateful to Joe Mandeville and Helen Deng as well as the Martinos Center radiopharmacy and imaging staff (Grae Arabasz, Shirley Hsu, Stephen Carlin, Chris Moseley, Nathan Schauer, Ehimen Aisaborhale, Judit Sore) for help with nonhuman primate experiments. HDAC activity screening was enabled by a Caliper assay developed by Jennifer Gale and Yan-Ling Zhang at the Broad Institute.
HDACs | histone deacetylases |
CNS | Center for Nanoscale Systems |
PET | positron emission tomography |
PPB | plasma protein binding |
DAT | dopamine transporter |
P-gp | P-glycoprotein |
NHP | nonhuman primate |
GWAS | genome-wide association studies |
SNPs | single nucleotide polymorphisms |
tPSA | total polar surface area |
TAC | time–activity curve |
References
This article references 45 other publications.
- 1Wang, W. Y.; Pan, L.; Su, S. C.; Quinn, E. J.; Sasaki, M.; Jimenez, J. C.; Mackenzie, I. R.; Huang, E. J.; Tsai, L. H. Interaction of FUS and HDAC1 regulates DNA damage response and repair in neurons Nat. Neurosci. 2013, 16, 1383– 1391Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhsVertbzI&md5=9849ff952f2ebb722f188fb9eba5c740Interaction of FUS and HDAC1 regulates DNA damage response and repair in neuronsWang, Wen-Yuan; Pan, Ling; Su, Susan C.; Quinn, Emma J.; Sasaki, Megumi; Jimenez, Jessica C.; MacKenzie, Ian R. A.; Huang, Eric J.; Tsai, Li-HueiNature Neuroscience (2013), 16 (10), 1383-1391CODEN: NANEFN; ISSN:1097-6256. (Nature Publishing Group)Defects in DNA repair have been extensively linked to neurodegenerative diseases, but the exact mechanisms remain poorly understood. We found that FUS, an RNA/DNA-binding protein that has been linked to amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration, is important for the DNA damage response (DDR). The function of FUS in DDR involved a direct interaction with histone deacetylase 1 (HDAC1), and the recruitment of FUS to double-stranded break sites was important for proper DDR signaling. Notably, FUS proteins carrying familial ALS mutations were defective in DDR and DNA repair and showed a diminished interaction with HDAC1. Moreover, we obsd. increased DNA damage in human ALS patients harboring FUS mutations. Our findings suggest that an impaired DDR and DNA repair may contribute to the pathogenesis of neurodegenerative diseases linked to FUS mutations.
- 2Bardai, F. H.; Verma, P.; Smith, C.; Rawat, V.; Wang, L.; D’Mello, S. R. Disassociation of histone deacetylase-3 from normal huntingtin underlies mutant huntingtin neurotoxicity J. Neurosci. 2013, 33, 11833– 11838Google Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhtFKqu77F&md5=f5d39b1df5ef060d99bf39d9bdb28a2cDisassociation of histone deacetylase-3 from normal Huntingtin underlies mutant huntingtin neurotoxicityBardai, Farah H.; Verma, Pragya; Smith, Chad; Rawat, Varun; Wang, Lulu; D'Mello, Santosh R.Journal of Neuroscience (2013), 33 (29), 11833-11838CODEN: JNRSDS; ISSN:0270-6474. (Society for Neuroscience)Huntington's disease (HD) is caused by a polyglutamine expansion within the huntingtin (Htt) protein. Both loss of function of normal Htt and gain of a toxic function by the polyglutamine-expanded mutant Htt protein have been proposed to be responsible for HD, although the mol. mechanisms involved are unclear. We show that Htt is a neuroprotective protein in both HD-related and unrelated model systems. Neuroprotection by Htt is mediated by its sequestration of histone deacetylase-3 (HDAC3), a protein known to promote neuronal death. In contrast to the normal Htt, mutant Htt interacts poorly with HDAC3. However, expression of mutant Htt liberates HDAC3 from Htt, thus de-repressing its neurotoxic activity. Indeed, mutant Htt neurotoxicity is inhibited by the knockdown of HDAC3 and markedly reduced in HDAC3-deficient neurons. A redn. in Htt-HDAC3 interaction is also seen in neurons exposed to other apoptotic stimuli and in the striatum of R6/2 HD mice. Our results suggest that the robust interaction between Htt and HDAC3 along with the ability of mutant Htt to disrupt this assocn. while not itself interacting with HDAC3 provides an explanation for both the loss-of-function and gain-of-toxic-function mechanisms proposed for HD. Moreover, our results identify HDAC3 as an essential player in mutant Htt-induced neurodegeneration.
- 3Bardai, F. H.; Price, V.; Zaayman, M.; Wang, L.; D’Mello, S. R. Histone deacetylase-1 (HDAC1) is a molecular switch between neuronal survival and death J. Biol. Chem. 2012, 287, 35444– 35453Google ScholarThere is no corresponding record for this reference.
- 4Graff, J.; Rei, D.; Guan, J. S.; Wang, W. Y.; Seo, J.; Hennig, K. M.; Nieland, T. J.; Fass, D. M.; Kao, P. F.; Kahn, M.; Su, S. C.; Samiei, A.; Joseph, N.; Haggarty, S. J.; Delalle, I.; Tsai, L. H. An epigenetic blockade of cognitive functions in the neurodegenerating brain Nature 2012, 483, 222– 226Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC383ptlCjtw%253D%253D&md5=6e567f82c587972acbdf75fbb06d2954An epigenetic blockade of cognitive functions in the neurodegenerating brainGraff Johannes; Rei Damien; Guan Ji-Song; Wang Wen-Yuan; Seo Jinsoo; Hennig Krista M; Nieland Thomas J F; Fass Daniel M; Kao Patricia F; Kahn Martin; Su Susan C; Samiei Alireza; Joseph Nadine; Haggarty Stephen J; Delalle Ivana; Tsai Li-HueiNature (2012), 483 (7388), 222-6 ISSN:.Cognitive decline is a debilitating feature of most neurodegenerative diseases of the central nervous system, including Alzheimer's disease. The causes leading to such impairment are only poorly understood and effective treatments are slow to emerge. Here we show that cognitive capacities in the neurodegenerating brain are constrained by an epigenetic blockade of gene transcription that is potentially reversible. This blockade is mediated by histone deacetylase 2, which is increased by Alzheimer's-disease-related neurotoxic insults in vitro, in two mouse models of neurodegeneration and in patients with Alzheimer's disease. Histone deacetylase 2 associates with and reduces the histone acetylation of genes important for learning and memory, which show a concomitant decrease in expression. Importantly, reversing the build-up of histone deacetylase 2 by short-hairpin-RNA-mediated knockdown unlocks the repression of these genes, reinstates structural and synaptic plasticity, and abolishes neurodegeneration-associated memory impairments. These findings advocate for the development of selective inhibitors of histone deacetylase 2 and suggest that cognitive capacities following neurodegeneration are not entirely lost, but merely impaired by this epigenetic blockade.
- 5Zhang, L.; Sheng, S.; Qin, C. The role of HDAC6 in Alzheimer’s disease J. Alzheimer’s Dis. 2013, 33, 283– 295Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhvVeksrbJ&md5=23363ee9d90e437ce4a8a017cd05c87eThe Role of HDAC6 in Alzheimer's DiseaseZhang, Ling; Sheng, Shuli; Qin, ChuanJournal of Alzheimer's Disease (2013), 33 (2), 283-295CODEN: JADIF9; ISSN:1387-2877. (IOS Press)A review. The expression of histone deacetylase 6 (HDAC6)-a versatile enzyme with a known role in epigenetics-increases significantly in the hippocampus and other relevant brain regions in both patients with Alzheimer's disease (AD) and animal models of AD. However, when and how HDAC6 expression increases during the course of AD progression remains unclear. Whether HDAC6 overexpression is an underlying cause of AD or a condition resulting from AD is controversial. Mounting evidence suggests that increased HDAC6 expression contributes to AD-assocd. neurodegeneration, although beneficial effects have also been identified. This review article addresses recent research on HDAC6 structure and function, and highlights the potential detrimental and protective roles of HDAC6 overexpression in AD. We hope to shed light on whether HDAC6 overexpression is assocd. with AD etiopathogenesis or whether it rescues AD-assocd. neurodegeneration compensatorily. Furthermore, we discuss new evidence showing that HDAC6 may be a therapeutic target for AD.
- 6Jakovcevski, M.; Bharadwaj, R.; Straubhaar, J.; Gao, G.; Gavin, D. P.; Jakovcevski, I.; Mitchell, A. C.; Akbarian, S. Prefrontal cortical dysfunction after overexpression of histone deacetylase 1 Biol. Psychiatry 2013, 74, 696– 705Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXnt1agu7g%253D&md5=88a220ddd46e10f52aa775eda5a8c761Prefrontal Cortical Dysfunction After Overexpression of Histone Deacetylase 1Jakovcevski, Mira; Bharadwaj, Rahul; Straubhaar, Juerg; Gao, Guangping; Gavin, David P.; Jakovcevski, Igor; Mitchell, Amanda C.; Akbarian, SchahramBiological Psychiatry (2013), 74 (9), 696-705CODEN: BIPCBF; ISSN:0006-3223. (Elsevier)Background: Postmortem brain studies have shown that HDAC1-a lysine deacetylase with broad activity against histones and nonhistone proteins-is frequently expressed at increased levels in prefrontal cortex (PFC) of subjects diagnosed with schizophrenia and related disease. However, it remains unclear whether upregulated expression of Hdac1 in the PFC could affect cognition and behavior. Methods: Using adeno-assocd. virus, an Hdac1 transgene was expressed in young adult mouse PFC, followed by behavioral assays for working and long-term memory, repetitive activity, and response to novelty. Prefrontal cortex transcriptomes were profiled by microarray. Antipsychotic drug effects were explored in mice treated for 21 days with haloperidol or clozapine. Results: Hdac1 overexpression in PFC neurons and astrocytes resulted in robust impairments in working memory, increased repetitive behaviors, and abnormal locomotor response profiles in novel environments. Long-term memory remained intact. Over 300 transcripts showed subtle but significant changes in Hdac1-overexpressing PFC. Major histocompatibility complex class II (MHC II)-related transcripts, including HLA-DQA1/H2-Aa, HLA-DQB1/H2-Ab1, and HLA-DRB1/H2-Eb1, located in the chromosome 6p21.3-22.1 schizophrenia and bipolar disorder risk locus, were among the subset of genes with a more robust (>1.5-fold) down-regulation in expression. Hdac1 levels declined during the course of normal PFC development. Antipsychotic drug treatment, including the atypical clozapine, did not affect Hdac1 levels in PFC but induced expression of multiple MHC II transcripts. Conclusions: Excessive HDAC1 activity, due to developmental defects or other factors, is assocd. with behavioral alterations and dysregulated expression of MHC II and other gene transcripts in the PFC.
- 7Morris, M. J.; Mahgoub, M.; Na, E. S.; Pranav, H.; Monteggia, L. M. Loss of histone deacetylase 2 improves working memory and accelerates extinction learning J. Neurosci. 2013, 33, 6401– 6411Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhtl2jtrvJ&md5=5be28a6994f7c9170ffe8c2bd535fcabLoss of histone deacetylase 2 improves working memory and accelerates extinction learningMorris, Michael J.; Mahgoub, Melissa; Na, Elisa S.; Pranav, Heena; Monteggia, Lisa M.Journal of Neuroscience (2013), 33 (15), 6401-6411CODEN: JNRSDS; ISSN:0270-6474. (Society for Neuroscience)Histone acetylation and deacetylation can be dynamically regulated in response to environmental stimuli and play important roles in learning and memory. Pharmacol. inhibition of histone deacetylases (HDACs) improves performance in learning tasks; however, many of these classical agents are "pan-HDAC" inhibitors, and their use makes it difficult to det. the roles of specific HDACs in cognitive function. We took a genetic approach using mice lacking the class I HDACs, HDAC1 or HDAC2, in postmitotic forebrain neurons to investigate the specificity or functional redundancy of these HDACs in learning and synaptic plasticity. We show that selective knock-out of Hdac2 led to a robust acceleration of the extinction rate of conditioned fear responses and a conditioned taste aversion as well as enhanced performance in an attentional set-shifting task. Hdac2 knock-out had no impact on episodic memory or motor learning, suggesting that the effects are task-dependent, with the predominant impact of HDAC2 inhibition being an enhancement in an animal's ability to rapidly adapt its behavioral strategy as a result of changes in associative contingencies. Our results demonstrate that the loss of HDAC2 improves associative learning, with no effect in nonassociative learning tasks, suggesting a specific role for HDAC2 in particular types of learning. HDAC2 may be an intriguing target for cognitive and psychiatric disorders that are characterized by an inability to inhibit behavioral responsiveness to maladaptive or no longer relevant assocns.
- 8Cao, D. J.; Wang, Z. V.; Battiprolu, P. K.; Jiang, N.; Morales, C. R.; Kong, Y.; Rothermel, B. A.; Gillette, T. G.; Hill, J. A. Histone deacetylase (HDAC) inhibitors attenuate cardiac hypertrophy by suppressing autophagy Proc. Natl. Acad. Sci. U.S.A. 2011, 108, 4123– 4128Google ScholarThere is no corresponding record for this reference.
- 9Ferguson, B. S.; Harrison, B. C.; Jeong, M. Y.; Reid, B. G.; Wempe, M. F.; Wagner, F. F.; Holson, E. B.; McKinsey, T. A. Signal-dependent repression of DUSP5 by class I HDACs controls nuclear ERK activity and cardiomyocyte hypertrophy Proc. Natl. Acad. Sci. U.S.A. 2013, 110, 9806– 9811Google ScholarThere is no corresponding record for this reference.
- 10Barnes, P. J. Corticosteroid resistance in patients with asthma and chronic obstructive pulmonary disease J. Allergy. Clin. Immunol. 2013, 131, 636– 645Google Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhvFWmtrY%253D&md5=b9f464c23387722c42b85280cc106d07Corticosteroid resistance in patients with asthma and chronic obstructive pulmonary diseaseBarnes, Peter J.Journal of Allergy and Clinical Immunology (2013), 131 (3), 636-645CODEN: JACIBY; ISSN:0091-6749. (Elsevier)A review. Reduced responsiveness to the anti-inflammatory effects of corticosteroids is a major barrier to effective management of asthma in smokers and patients with severe asthma and in the majority of patients with chronic obstructive pulmonary disease (COPD). The mol. mechanisms leading to steroid resistance are now better understood, and this has identified new targets for therapy. In patients with severe asthma, several mol. mechanisms have been identified that might account for reduced steroid responsiveness, including reduced nuclear translocation of glucocorticoid receptor (GR) α after binding corticosteroids. This might be due to modification of the GR by means of phosphorylation as a result of activation of several kinases (p38 mitogen-activated protein kinase α, p38 mitogen-activated protein kinase γ, and c-Jun N-terminal kinase 1), which in turn might be due to reduced activity and expression of phosphatases, such as mitogen-activated protein kinase phosphatase 1 and protein phosphatase A2. Other mechanisms proposed include increased expression of GRβ, which competes with and thus inhibits activated GRα; increased secretion of macrophage migration inhibitory factor; competition with the transcription factor activator protein 1; and reduced expression of histone deacetylase (HDAC) 2. HDAC2 appears to mediate the action of steroids to switch off activated inflammatory genes, but in patients with COPD, patients with severe asthma, and smokers with asthma, HDAC2 activity and expression are reduced by oxidative stress through activation of phosphoinositide 3-kinase δ. Strategies for managing steroid resistance include alternative anti-inflammatory drugs, but a novel approach is to reverse steroid resistance by increasing HDAC2 expression, which can be achieved with theophylline and phosphoinositide 3-kinase δ inhibitors. Long-acting β2-agonists can also increase steroid responsiveness by reversing GRα phosphorylation. Identifying the mol. mechanisms of steroid resistance in asthmatic patients and patients with COPD can thus lead to more effective anti-inflammatory treatments.
- 11Brochier, C.; Dennis, G.; Rivieccio, M. A.; McLaughlin, K.; Coppola, G.; Ratan, R. R.; Langley, B. Specific acetylation of p53 by HDAC inhibition prevents DNA damage-induced apoptosis in neurons J. Neurosci. 2013, 33, 8621– 8632Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXotFant7w%253D&md5=1b5b2f3f53c0802a60ab93a9bf6bc537Specific acetylation of p53 by HDAC inhibition prevents DNA damage-induced apoptosis in neuronsBrochier, Camille; Dennis, Gretel; Rivieccio, Mark A.; McLaughlin, Kathryn; Coppola, Giovanni; Ratan, Rajiv R.; Langley, BrettJournal of Neuroscience (2013), 33 (20), 8621-8632CODEN: JNRSDS; ISSN:0270-6474. (Society for Neuroscience)Histone deacetylase (HDAC) inhibitors have been used to promote neuronal survival and ameliorate neurol. dysfunction in a host of neurodegenerative disease models. The precise mol. mechanisms whereby HDAC inhibitors prevent neuronal death are currently the focus of intensive research. Here we demonstrate that HDAC inhibition prevents DNA damage-induced neurodegeneration by modifying the acetylation pattern of the tumor suppressor p53, which decreases its DNA-binding and transcriptional activation of target genes. Specifically, we identify that acetylation at K382 and K381 prevents p53 from assocg. with the pro-apoptotic PUMA gene promoter, activating transcription, and inducing apoptosis in mouse primary cortical neurons. Paradoxically, acetylation of p53 at the same lysines in various cancer cell lines leads to the induction of PUMA expression and death. Together, our data provide a mol. understanding of the specific outcomes of HDAC inhibition and suggest that strategies aimed at enhancing p53 acetylation at K381 and K382 might be therapeutically viable for capturing the beneficial effects in the CNS, without compromising tumor suppression.
- 12Zhang, Z.; Yamashita, H.; Toyama, T.; Sugiura, H.; Omoto, Y.; Ando, Y.; Mita, K.; Hamaguchi, M.; Hayashi, S.; Iwase, H. HDAC6 expression is correlated with better survival in breast cancer Clin. Cancer Res. 2004, 10, 6962– 6968Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXpslGlur4%253D&md5=c04ab7b31de3132c37a18ed1d5ff54feHDAC6 expression is correlated with better survival in breast cancerZhang, Zhenhuan; Yamashita, Hiroko; Toyama, Tatsuya; Sugiura, Hiroshi; Omoto, Yoko; Ando, Yoshiaki; Mita, Keiko; Hamaguchi, Maho; Hayashi, Shin-ichi; Iwase, HirotakaClinical Cancer Research (2004), 10 (20), 6962-6968CODEN: CCREF4; ISSN:1078-0432. (American Association for Cancer Research)The structure and function of chromatin can be altered by modifications to histone. Histone acetylation in vivo is a dynamic reversible process governed by histone acetyltransferases (HATs) and histone deacetylases (HDACs). HDAC6 is a unique isoform among the HDACs, and a gene expression pattern study, with cDNA microarray in MCF-7 cells, showed the HDAC6 gene to be late responsive, estrogen induced, and up-regulated. This led us to hypothesize that there was a link between levels of HDAC6 expression and the metastatic potential of breast cancer and also, therefore, the prognosis of these patients. In the present study, the level of HDAC6 mRNA expression was analyzed with quant. real-time reverse transcription-PCR, in 135 female patients with invasive breast cancer. HDAC6 protein expression was also detd. by immunohistochem. An assocn. was sought between HDAC6 expression and various clinicopathol. factors. HDAC6 mRNA was expressed at significantly higher levels in breast cancer patients with small tumors measuring less than 2 cm, with low histol. grade, and in estrogen receptor α- and progesterone receptor-pos. tumors. By contrast, no relationship was found between HDAC6 mRNA expression and any of the other clinicopathol. factors, namely, age, menopausal status, and axillary lymph node involvement. Patients expressing high levels of HDAC6 mRNA and protein had a better prognosis than those expressing low levels, in terms of disease-free survival. However, multivariate anal. failed to show that HDAC6 mRNA and protein are an independent prognostic factors for disease-free survival and overall survival. Furthermore, the patients with high levels of HDAC6 mRNA tended to be more responsive to endocrine treatment than those with low levels. Specific HDAC6 staining was found in the nucleus of some normal epithelial cells and in the cytoplasm of the majority of cancer cells. Although postmenopausal patients showed higher HDAC6 protein expression, there were no relationship between protein expression and any other clinicopathol. factors. We conclude that the levels of HDAC6 mRNA expression may have potential both as a marker of endocrine responsiveness and also as a prognostic indicator in breast cancer. Addnl. investigations are warranted concerning the relationship between HDAC6 expression and response to endocrine therapy.
- 13Rajendran, P.; Kidane, A. I.; Yu, T. W.; Dashwood, W. M.; Bisson, W. H.; Lohr, C. V.; Ho, E.; Williams, D. E.; Dashwood, R. H. HDAC turnover, CtIP acetylation and dysregulated DNA damage signaling in colon cancer cells treated with sulforaphane and related dietary isothiocyanates Epigenetics 2013, 8, 612– 623Google ScholarThere is no corresponding record for this reference.
- 14Cai, Y.; Geutjes, E. J.; de Lint, K.; Roepman, P.; Bruurs, L.; Yu, L. R.; Wang, W.; van Blijswijk, J.; Mohammad, H.; de Rink, I.; Bernards, R.; Baylin, S. B. The NuRD complex cooperates with DNMTs to maintain silencing of key colorectal tumor suppressor genes Oncogene 2014, 33, 2157– 2168Google ScholarThere is no corresponding record for this reference.
- 15Kim, M. G.; Pak, J. H.; Choi, W. H.; Park, J. Y.; Nam, J. H.; Kim, J. H. The relationship between cisplatin resistance and histone deacetylase isoform overexpression in epithelial ovarian cancer cell lines J. Gynecol. Oncol. 2012, 23, 182– 189Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXmt1Sls7o%253D&md5=4086ae72007b8a38be1462349d4ab2d5The relationship between cisplatin resistance and histone deacetylase isoform overexpression in epithelial ovarian cancer cell linesKim, Min-Gyun; Pak, Jhang Ho; Choi, Won Ho; Park, Jeong-Yeol; Nam, Joo-Hyun; Kim, Jong-HyeokJournal of Gynecologic Oncology (2012), 23 (3), 182-189CODEN: JGOOAH; ISSN:2005-0380. (Korean Society of Gynecologic Oncology and Colposcopy)Objective: To investigate the relationship between cisplatin resistance and histone deacetylase (HDAC) isoform overexpression in ovarian cancer cell lines. Methods: Expression of four HDAC isoforms (HDAC 1, 2, 3, and 4) in two ovarian cancer cell lines, SKOV3 and OVCAR3, exposed to various concns. of cisplatin was examd. by western blot analyses. Cells were transfected with plasmid DNA of each HDAC. The overexpression of protein and mRNA of each HDAC was confirmed by western blot and reverse transcriptase-polymerase chain reaction analyses, resp. The cell viability of the SKOV3 and OVCAR3 cells transfected with HDAC plasmid DNA was measured using the cell counting kit-8 assay after treatment with cisplatin. Results: The 50% inhibitory concn. of the SKOV3 and OVCAR3 cells can be detd. 15-24 h after treatment with 15 μg/mL cisplatin. The expression level of acetylated histone 3 protein in SKOV3 cells increased after exposure to cisplatin. Compared with control cells at 24 h after cisplatin exposure, the viability of SKOV3 cells overexpressing HDAC 1 and 3 increased by 15% and 13% (p<0.05), resp. On the other hand, OVCAR3 cells that overexpressed HDAC 2 and 4 exhibited increased cell viability by 23% and 20% (p<0.05), resp., compared with control cells 24 h after exposure to cisplatin. Conclusion: In SKOV3 and OVCAR3 epithelial ovarian cancer cell lines, the correlation between HDAC overexpression and cisplatin resistance was confirmed. However, the specific HDAC isoform assocd. with resistance to cisplatin varied depending on the ovarian cancer cell line. These results may suggest that each HDAC isoform conveys cisplatin resistance via different mechanisms.
- 16Hayashi, A.; Horiuchi, A.; Kikuchi, N.; Hayashi, T.; Fuseya, C.; Suzuki, A.; Konishi, I.; Shiozawa, T. Type-specific roles of histone deacetylase (HDAC) overexpression in ovarian carcinoma: HDAC1 enhances cell proliferation and HDAC3 stimulates cell migration with downregulation of E-cadherin Int. J. Cancer. 2010, 127, 1332– 1346Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXpt1Ohsbo%253D&md5=35c0d48c3512ea474029d0ad80956c4dType-specific roles of histone deacetylase (HDAC) overexpression in ovarian carcinoma: HDAC1 enhances cell proliferation and HDAC3 stimulates cell migration with downregulation of E-cadherinHayashi, Akiko; Horiuchi, Akiko; Kikuchi, Norihiko; Hayashi, Takuma; Fuseya, Chiho; Suzuki, Akihisa; Konishi, Ikuo; Shiozawa, TanriInternational Journal of Cancer (2010), 127 (6), 1332-1346CODEN: IJCNAW; ISSN:0020-7136. (Wiley-Liss, Inc.)Histone acetylation/deacetylation controls chromatin activity and subsequent gene transcription. Recent studies demonstrated the activation of histone deacetylases (HDACs) in various human malignancies; however, the expression and function of HDACs in ovarian tumors are not fully understood. In this study, we examd. the immunohistochem. expression of HDAC1, HDAC2 and HDAC3 using tissues obtained from 115 cases of ovarian tumors and compared it with that of Ki-67 (a growth marker), p21, and E-cadherin and clinicopathol. parameters. In addn., we analyzed the effect of specific siRNA for HDAC1, HDAC2 and HDAC3 on the expression of cell cycle-related mols. and E-cadherin to clarify the functional difference among the 3 HDACs. The results indicated that the immunohistochem. expression of nuclear HDAC1, HDAC2 and HDAC3 proteins increased stepwise in benign, borderline and malignant tumors. The expression of HDAC1 and HDAC2 was correlated with Ki-67 expression and that of HDAC3 was inversely correlated with E-cadherin expression. Among the HDACs examd., only HDAC1 was assocd. with a poor outcome, when overexpressed. Treatment with HDAC inhibitors suppressed the proliferation of ovarian cancer cells in assocn. with apoptosis. A specific siRNA for HDAC1 significantly reduced the proliferation of ovarian carcinoma cells via downregulation of cyclin A expression, but siRNA for HDAC3 reduced the cell migration with elevated E-cadherin expression. Our results suggested that HDAC1 plays an important role in the proliferation of ovarian cancer cells, whereas HDAC3 functions in cell adhesion and migration. Therefore, specific therapeutic approaches should be considered according to the HDAC subtypes.
- 17Benes, F. M.; Lim, B.; Matzilevich, D.; Walsh, J. P.; Subburaju, S.; Minns, M. Regulation of the GABA cell phenotype in hippocampus of schizophrenics and bipolars Proc. Natl. Acad. Sci. U.S.A. 2007, 104, 10164– 10169Google ScholarThere is no corresponding record for this reference.
- 18Broide, R. S.; Redwine, J. M.; Aftahi, N.; Young, W.; Bloom, F. E.; Winrow, C. J. Distribution of histone deacetylases 1–11 in the rat brain J. Mol. Neurosci. 2007, 31, 47– 58Google Scholar18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXls1GqtL8%253D&md5=8b0ef986705015f17dee856b602a99eaDistribution of histone deacetylases 1-11 in the rat brainBroide, Ron S.; Redwine, Jeff M.; Aftahi, Najla; Young, Warren; Bloom, Floyd E.; Winrow, Christopher J.Journal of Molecular Neuroscience (2007), 31 (1), 47-58CODEN: JMNEES; ISSN:0895-8696. (Humana Press Inc.)Although protein phosphorylation has been characterized more extensively, modulation of the acetylation state of signaling mols. is now being recognized as a key means of signal transduction. The enzymes responsible for mediating these changes include histone acetyltransferases and histone deacetylases (HDACs). Members of the HDAC family of enzymes have been identified as potential therapeutic targets for diseases ranging from cancer to ischemia and neurodegeneration. The authors initiated a project to conduct comprehensive gene expression mapping of the 11 HDAC isoforms (HDAC1-11) (classes I, II, and IV) throughout the rat brain using high-resoln. in situ hybridization (ISH) and imaging technol. Internal and external data bases were employed to identify the appropriate rat sequence information for probe selection. In addn., immunohistochem. was performed on these samples to sep. examine HDAC expression in neurons, astrocytes, oligodendrocytes, and endothelial cells in the CNS. This double-labeling approach enabled the identification of specific cell types in which the individual HDACs were expressed. The signals obtained by ISH were compared to radiolabeled stds. and thereby enabled semiquant. anal. of individual HDAC isoforms and defined relative levels of gene expression in >50 brain regions. This project produced an extensive atlas of 11 HDAC isoforms throughout the rat brain, including cell type localization, providing a valuable resource for examg. the roles of specific HDACs in the brain and the development of future modulators of HDAC activity.
- 19Hooker, J. M.; Kim, S. W.; Alexoff, D.; Xu, Y.; Shea, C.; Reid, A.; Volkow, N.; Fowler, J. S. Histone deacetylase inhibitor, MS-275, exhibits poor brain penetration: PK studies of [11C]MS-275 using Positron Emission Tomography ACS Chem. Neurosci. 2010, 1, 65– 73Google Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhtlKms7vL&md5=f1cf8e753ac80029fae6cf77193f172fHistone Deacetylase Inhibitor MS-275 Exhibits Poor Brain Penetration: Pharmacokinetic Studies of [11C]MS-275 using Positron Emission TomographyHooker, Jacob M.; Kim, Sung Won; Alexoff, David; Xu, Youwen; Shea, Colleen; Reid, Alicia; Volkow, Nora; Fowler, Joanna S.ACS Chemical Neuroscience (2010), 1 (1), 65-73CODEN: ACNCDM; ISSN:1948-7193. (American Chemical Society)MS-275 (entinostat) is a histone deacetylase (HDAC) inhibitor currently in clin. trials for the treatment of several types of cancer. Recent reports have noted that MS-275 can cross the blood-brain barrier (BBB) and cause region-specific changes in rodent brain histone acetylation. To characterize the pharmacokinetics and distribution of MS-275 in the brain using positron emission tomog. (PET), we labeled the carbamate carbon of MS-275 with carbon-11. Using PET, we detd. that [11C]MS-275 has low uptake in brain tissue when administered i.v. to nonhuman primates. In rodent studies, we obsd. that pharmacokinetics and brain accumulation of [11C]MS-275 were not changed by the coadministration of large doses of unlabeled MS-275. These results, which both highlight the poor brain penetration of MS-275, clearly suggest its limitation as a therapeutic agent for the central nervous system (CNS). Moreover, our study demonstrates the effectiveness of PET at providing brain pharmacokinetic data for HDAC inhibitors. These data are important not only for the development of new compds. for peripheral cancer treatment (where CNS exclusion is often advantageous) but also for the treatment of neurol. disorders (where CNS penetration is crit.).
- 20Yeh, H. H.; Tian, M.; Hinz, R.; Young, D.; Shavrin, A.; Mukhapadhyay, U.; Flores, L. G.; Balatoni, J.; Soghomonyan, S.; Jeong, H. J.; Pal, A.; Uthamanthil, R.; Jackson, J. N.; Nishi, R.; Mizuma, H.; Onoe, H.; Kagawa, S.; Higashi, T.; Fukumitsu, N.; Alauddin, M.; Tong, W.; Herholz, K.; Gelovani, J. G. Imaging epigenetic regulation by histone deacetylases in the brain using PET/MRI with (1)(8)F-FAHA Neuroimage 2013, 64, 630– 639Google ScholarThere is no corresponding record for this reference.
- 21Hendricks, J. A.; Keliher, E. J.; Marinelli, B.; Reiner, T.; Weissleder, R.; Mazitschek, R. In vivo PET imaging of histone deacetylases by 18F-suberoylanilide hydroxamic acid (18F-SAHA) J. Med. Chem. 2011, 54, 5576– 5582Google ScholarThere is no corresponding record for this reference.
- 22Seo, Y. J.; Kang, Y.; Muench, L.; Reid, A.; Caesar, S.; Jean, L.; Fevier-Wagner, F.; Holson, E. B.; Haggarty, S. J.; Weiss, P.; King, P.; Carter, P.; Volkow, N. D.; Fowler, J. S.; Hooker, J. M.; Kim, S. W. Image-guided synthesis reveals potent blood-brain barrier permeable histone deacetylase inhibitors ACS Chem. Neurosci. 2014, 5, 588– 596Google Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXmvF2msb4%253D&md5=c59102403f07049034e1cd0a47161eb6Image-Guided Synthesis Reveals Potent Blood-Brain Barrier Permeable Histone Deacetylase InhibitorsSeo, Young Jun; Kang, Yeona; Muench, Lisa; Reid, Alicia; Caesar, Shannon; Jean, Logan; Wagner, Florence; Holson, Edward; Haggarty, Stephen J.; Weiss, Philipp; King, Payton; Carter, Pauline; Volkow, Nora D.; Fowler, Joanna S.; Hooker, Jacob M.; Kim, Sung WonACS Chemical Neuroscience (2014), 5 (7), 588-596CODEN: ACNCDM; ISSN:1948-7193. (American Chemical Society)Recent studies have revealed that several histone deacetylase (HDAC) inhibitors, which are used to study/treat brain diseases, show low blood-brain barrier (BBB) penetration. In addn. to low HDAC potency and selectivity obsd., poor brain penetrance may account for the high doses needed to achieve therapeutic efficacy. Here the authors report the development and evaluation of highly potent and blood-brain barrier permeable HDAC inhibitors for CNS applications based on an image-guided approach involving the parallel synthesis and radiolabeling of a series of compds. based on the benzamide HDAC inhibitor, MS-275 as a template. BBB penetration was optimized by rapid carbon-11 labeling and PET imaging in the baboon model and using the imaging derived data on BBB penetration from each compd. to feed back into the design process. A total of 17 compds. were evaluated, revealing mols. with both high binding affinity and BBB permeability. A key element conferring BBB penetration in this benzamide series was a basic benzylic amine. These derivs. exhibited 1-100 nM inhibitory activity against recombinant human HDAC1 and HDAC2. Three of the carbon-11 labeled aminomethyl benzamide derivs. showed high BBB penetration (∼0.015%ID/cc) and regional binding heterogeneity in the brain (high in thalamus and cerebellum). Taken together this approach has afforded a strategy and a predictive model for developing highly potent and BBB permeable HDAC inhibitors for CNS applications and for the discovery of novel candidate mols. for small mol. probes and drugs.
- 23Seo, Y. J.; Muench, L.; Reid, A.; Chen, J.; Kang, Y.; Hooker, J. M.; Volkow, N. D.; Fowler, J. S.; Kim, S. W. Radionuclide labeling and evaluation of candidate radioligands for PET imaging of histone deacetylase in the brain Bioorg. Med. Chem. Lett. 2013, 23, 6700– 6705Google ScholarThere is no corresponding record for this reference.
- 24Hanson, J. E.; La, H.; Plise, E.; Chen, Y. H.; Ding, X.; Hanania, T.; Sabath, E. V.; Alexandrov, V.; Brunner, D.; Leahy, E.; Steiner, P.; Liu, L.; Scearce-Levie, K.; Zhou, Q. SAHA enhances synaptic function and plasticity in vitro but has limited brain availability in vivo and does not impact cognition PLoS One 2013, 8, e69964Google ScholarThere is no corresponding record for this reference.
- 25Maugh, T. H., 2nd Panel urges wide use of antiviral drug Science 1979, 206, 1058– 1060Google ScholarThere is no corresponding record for this reference.
- 26Banister, S. D.; Wilkinson, S. M.; Longworth, M.; Stuart, J.; Apetz, N.; English, K.; Brooker, L.; Goebel, C.; Hibbs, D. E.; Glass, M.; Connor, M.; McGregor, I. S.; Kassiou, M. The synthesis and pharmacological evaluation of adamantane-derived indoles: cannabimimetic drugs of abuse ACS Chem. Neurosci. 2013, 4, 1081– 1092Google Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXltV2nu7c%253D&md5=859bff2f95ad6a0931b013ac532b3407The Synthesis and Pharmacological Evaluation of Adamantane-Derived Indoles: Cannabimimetic Drugs of AbuseBanister, Samuel D.; Wilkinson, Shane M.; Longworth, Mitchell; Stuart, Jordyn; Apetz, Nadine; English, Katrina; Brooker, Lance; Goebel, Catrin; Hibbs, David E.; Glass, Michelle; Connor, Mark; McGregor, Iain S.; Kassiou, MichaelACS Chemical Neuroscience (2013), 4 (7), 1081-1092CODEN: ACNCDM; ISSN:1948-7193. (American Chemical Society)Two novel adamantane derivs., adamantan-1-yl(1-pentyl-1H-indol-3-yl)methanone (AB-001) and N-(adamtan-1-yl)-1-pentyl-1H-indole-3-carboxamide (SDB-001), were recently identified as cannabimimetic indoles of abuse. Conflicting anecdotal reports of the psychoactivity of AB-001 in humans, and a complete dearth of information about the bioactivity of SDB-001, prompted the prepn. of AB-001, SDB-001, and several analogs intended to explore preliminary structure-activity relationships within this class. This study sought to elucidate which structural features of AB-001, SDB-001, and their analogs govern the cannabimimetic potency of these chemotypes in vitro and in vivo. All compds. showed similar full agonist profiles at CB1 (EC50 = 16-43 nM) and CB2 (EC50 = 29-216 nM) receptors in vitro using a FLIPR membrane potential assay, with the exception of SDB-002, which demonstrated partial agonist activity at CB2 receptors. The activity of AB-001, AB-002, and SDB-001 in rats was compared to that of Δ9-tetrahydrocannabinol (Δ9-THC) and cannabimimetic indole JWH-018 using biotelemetry. SDB-001 dose-dependently induced hypothermia and reduced heart rate (maximal dose 10 mg/kg) with potency comparable to that of Δ9-tetrahydrocannabinol (Δ9-THC, maximal dose 10 mg/kg), and lower than that of JWH-018 (maximal dose 3 mg/kg). Addnl., the changes in body temp. and heart rate affected by SDB-001 are of longer duration than those of Δ9-THC or JWH-018, suggesting a different pharmacokinetic profile. In contrast, AB-001, and its homolog, AB-002, did not produce significant hypothermic and bradycardic effects, even at relatively higher doses (up to 30 mg/kg), indicating greatly reduced potency compared to Δ9-THC, JWH-018, and SDB-001.
- 27Gopalan, B.; Ponpandian, T.; Kachhadia, V.; Bharathimohan, K.; Vignesh, R.; Sivasudar, V.; Narayanan, S.; Mandar, B.; Praveen, R.; Saranya, N.; Rajagopal, S. Discovery of adamantane based highly potent HDAC inhibitors Bioorg. Med. Chem. Lett. 2013, 23, 2532– 2537Google ScholarThere is no corresponding record for this reference.
- 28Farde, L.; Eriksson, L.; Blomquist, G.; Halldin, C. Kinetic analysis of central [11C]raclopride binding to D2-dopamine receptors studied by PET--a comparison to the equilibrium analysis J. Cereb. Blood Flow Metab. 1989, 9, 696– 708Google Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL1MXmtV2msLY%253D&md5=86b7f5f4a1d44222f2009990c889b1b7Kinetic analysis of central [11C]raclopride binding to D2-dopamine receptors studied by PET - a comparison to the equilibrium analysisFarde, L.; Eriksson, L.; Blomquist, G.; Halldin, C.Journal of Cerebral Blood Flow and Metabolism (1989), 9 (5), 696-708CODEN: JCBMDN; ISSN:0271-678X.[11C]raclopride binding to central D2-dopamine receptors in humans has previously been examd. by positron emission tomog. (PET). Based on the rapid occurrence of binding equil., a satn. anal. was developed for the detn. of receptor d. (Bmax) and affinity (Kd). For anal. of PET measurements obtained with other ligands, a kinetic 3-compartment model was used. In the present study, the brain uptake of [11C]raclopride was analyzed further by applying both a kinetic and an equil. anal. to data obtained from 4 PET expts. in each of 3 healthy subjects. First regional cerebral blood vol. was detd. In the second and third expt., [11C]raclopride with high and low specific activity was used. In a fourth expt., the [11C]raclopride enantiomer [11C]FLB472 was used to examine the concn. of free radioligand and nonspecific binding in brain. Radioactivity in arterial blood was measured using an automated blood sampling system. Bmax And Kd values for [11C]raclopride binding could be detd. also with the kinetic anal. As expected theor., those values were similar to those obtained with the equil. anal. In addn., the kinetic anal. allowed sep. detn. of the assocn. and dissocn. rate consts., kon and koff, resp. Examn. of [11C]raclopride and [11C]FLB472 uptake in brain regions devoid of specific D2-dopamine receptor binding indicated a fourth compartment in which uptake was reversible, nonstereoselective, and nonsaturable in the dose range studied.
- 29Innis, R. B.; Cunningham, V. J.; Delforge, J.; Fujita, M.; Gjedde, A.; Gunn, R. N.; Holden, J.; Houle, S.; Huang, S. C.; Ichise, M.; Iida, H.; Ito, H.; Kimura, Y.; Koeppe, R. A.; Knudsen, G. M.; Knuuti, J.; Lammertsma, A. A.; Laruelle, M.; Logan, J.; Maguire, R. P.; Mintun, M. A.; Morris, E. D.; Parsey, R.; Price, J. C.; Slifstein, M.; Sossi, V.; Suhara, T.; Votaw, J. R.; Wong, D. F.; Carson, R. E. Consensus nomenclature for in vivo imaging of reversibly binding radioligands J. Cereb. Blood Flow Metab. 2007, 27, 1533– 1539Google Scholar29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXptlSqtrc%253D&md5=77f7e875203e18e1a15d3e2d7fcd4ac6Consensus nomenclature for in vivo imaging of reversibly binding radioligandsInnis, Robert B.; Cunningham, Vincent J.; Delforge, Jacques; Fujita, Masahiro; Gjedde, Albert; Gunn, Roger N.; Holden, James; Houle, Sylvain; Huang, Sung-Cheng; Ichise, Masanori; Iida, Hidehiro; Ito, Hiroshi; Kimura, Yuichi; Koeppe, Robert A.; Knudsen, Gitte M.; Knuuti, Juhani; Lammertsma, Adriaan A.; Laruelle, Marc; Logan, Jean; Maguire, Ralph Paul; Mintun, Mark A.; Morris, Evan D.; Parsey, Ramin; Price, Julie C.; Slifstein, Mark; Sossi, Vesna; Suhara, Tetsuya; Votaw, John R.; Wong, Dean F.; Carson, Richard E.Journal of Cerebral Blood Flow & Metabolism (2007), 27 (9), 1533-1539CODEN: JCBMDN; ISSN:0271-678X. (Nature Publishing Group)A review. An international group of experts in pharmacokinetic modeling recommends a consensus nomenclature to describe in vivo mol. imaging of reversibly binding radioligands.
- 30Wang, Y.; Zhang, Y. L.; Hennig, K.; Gale, J. P.; Hong, Y.; Cha, A.; Riley, M.; Wagner, F.; Haggarty, S. J.; Holson, E.; Hooker, J. Class I HDAC imaging using [ (3)H]CI-994 autoradiography Epigenetics 2013, 8, 756– 764Google ScholarThere is no corresponding record for this reference.
- 31Schroeder, F. A.; Lewis, M. C.; Fass, D. M.; Wagner, F. F.; Zhang, Y. L.; Hennig, K. M.; Gale, J.; Zhao, W. N.; Reis, S.; Barker, D. D.; Berry-Scott, E.; Kim, S. W.; Clore, E. L.; Hooker, J. M.; Holson, E. B.; Haggarty, S. J.; Petryshen, T. L. A selective HDAC 1/2 inhibitor modulates chromatin and gene expression in brain and alters mouse behavior in two mood-related tests PLoS One 2013, 8, e71323Google ScholarThere is no corresponding record for this reference.
- 32Fass, D. M.; Schroeder, F. A.; Perlis, R. H.; Haggarty, S. J. Epigenetic mechanisms in mood disorders: Targeting neuroplasticity Neuroscience 2014, 264, 112– 130Google ScholarThere is no corresponding record for this reference.
- 33Davies, G.; Tenesa, A.; Payton, A.; Yang, J.; Harris, S. E.; Liewald, D.; Ke, X.; Le Hellard, S.; Christoforou, A.; Luciano, M.; McGhee, K.; Lopez, L.; Gow, A. J.; Corley, J.; Redmond, P.; Fox, H. C.; Haggarty, P.; Whalley, L. J.; McNeill, G.; Goddard, M. E.; Espeseth, T.; Lundervold, A. J.; Reinvang, I.; Pickles, A.; Steen, V. M.; Ollier, W.; Porteous, D. J.; Horan, M.; Starr, J. M.; Pendleton, N.; Visscher, P. M.; Deary, I. J. Genome-wide association studies establish that human intelligence is highly heritable and polygenic Mol. Psychiatry 2011, 16, 996– 1005Google Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXht1CltrbM&md5=0287370fcfc4ced4dbe3d07b982cdcf0Genome-wide association studies establish that human intelligence is highly heritable and polygenicDavies, G.; Tenesa, A.; Payton, A.; Yang, J.; Harris, S. E.; Liewald, D.; Ke, X.; Le Hellard, S.; Christoforou, A.; Luciano, M.; McGhee, K.; Lopez, L.; Gow, A. J.; Corley, J.; Redmond, P.; Fox, H. C.; Haggarty, P.; Whalley, L. J.; McNeill, G.; Goddard, M. E.; Espeseth, T.; Lundervold, A. J.; Reinvang, I.; Pickles, A.; Steen, V. M.; Ollier, W.; Porteous, D. J.; Horan, M.; Starr, J. M.; Pendleton, N.; Visscher, P. M.; Deary, I. J.Molecular Psychiatry (2011), 16 (10), 996-1005CODEN: MOPSFQ; ISSN:1359-4184. (Nature Publishing Group)General intelligence is an important human quant. trait that accounts for much of the variation in diverse cognitive abilities. Individual differences in intelligence are strongly assocd. with many important life outcomes, including educational and occupational attainments, income, health and lifespan. Data from twin and family studies are consistent with a high heritability of intelligence, but this inference has been controversial. The authors conducted a genome-wide anal. of 3511 unrelated adults with data on 549 692 single nucleotide polymorphisms (SNPs) and detailed phenotypes on cognitive traits. They est. that 40% of the variation in crystd.-type intelligence and 51% of the variation in fluid-type intelligence between individuals is accounted for by linkage disequil. between genotyped common SNP markers and unknown causal variants. These ests. provide lower bounds for the narrow-sense heritability of the traits. The authors partitioned genetic variation on individual chromosomes and found that, on av., longer chromosomes explain more variation. Finally, using just SNP data the authors predicted ∼1% of the variance of crystd. and fluid cognitive phenotypes in an independent sample (P=0.009 and 0.028, resp.). These results unequivocally confirm that a substantial proportion of individual differences in human intelligence is due to genetic variation, and are consistent with many genes of small effects underlying the additive genetic influences on intelligence. Mol. Psychiatry (2011) 16, 996-1005; doi:10.1038/mp.2011.85; published online 9 August 2011.
- 34McPherson, R. From genome-wide association studies to functional genomics: new insights into cardiovascular disease Can. J. Cardiol. 2013, 29, 23– 29Google Scholar34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC3s7otVelug%253D%253D&md5=87afdc14bea7fcb43daaa8fdde3ea0ebFrom genome-wide association studies to functional genomics: new insights into cardiovascular diseaseMcPherson RuthThe Canadian journal of cardiology (2013), 29 (1), 23-9 ISSN:.Genome-wide association studies (GWASs) for coronary artery disease (CAD) have identified more than 30 variants robustly associated with CAD risk. The majority are not associated with conventional risk factors but highlight novel pathways, including cellular proliferation. Although some risk variants are nonsynonymous coding variants resulting in an amino acid change in the encoded protein, the majority are in noncoding regions of the genome and may encompass multiple signals of variable effect. The use of genetic data for development of new therapies requires the identification of causative genetic variants and elucidation of the molecular mechanisms by which they predispose to CAD. The computational and laboratory approaches for the interpretation of GWAS data are discussed with a particular focus on noncoding variants, including the study of regulatory elements, the evaluation of nonsynonymous coding variants, and expression quantitative trait locus analysis for the integration of GWAS data with genome-wide messenger RNA expression data.
- 35Torres, J. M.; Cox, N. J.; Philipson, L. H. Genome wide association studies for diabetes: perspective on results and challenges Pediatr. Diabetes 2013, 14, 90– 96Google Scholar35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXmvFKnsrY%253D&md5=300744395e3830b6b92d359a4516da28Genome wide association studies for diabetes: perspective on results and challengesTorres, J. M.; Cox, N. J.; Philipson, L. H.Pediatric Diabetes (2013), 14 (2), 90-96CODEN: PDEIBT; ISSN:1399-5448. (Wiley-Blackwell)A review. Recent results of genome wide assocn. study (GWAS) for diabetes genes, while reaching impressive tech. milestones and implicating new findings for research, have been uniformly disappointing in terms of immediate clin. utility. The relative risk assocd. with any of the newly reported genetic loci, or even considering all of them together, is far less than simply that which can be obtained by taking a history and a phys. exam. For type 2 diabetes (T2D), GWAS have implicated novel pathways, supported previously known assocns., and highlighted the importance of the beta cell and insulin secretion. Monogenic forms of diabetes, on the other hand, continue to yield interesting insights into genes controlling human beta cell function but most cases of monogenic diabetes are simply not diagnosed. Here, we briefly review recent results related to type 1, type 2 and maturity onset diabetes of youth (MODY) diabetes and suggest that future studies emphasizing quant. traits are likely to yield even more insights.
- 36Mells, G. F.; Kaser, A.; Karlsen, T. H. Novel insights into autoimmune liver diseases provided by genome-wide association studies J. Autoimmun. 2013, 46, 41– 54Google Scholar36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXht1eksLjL&md5=bc5b8b336c89d14c3c2a1f25ba1e0217Novel insights into autoimmune liver diseases provided by genome-wide association studiesMells, George F.; Kaser, Arthur; Karlsen, Tom H.Journal of Autoimmunity (2013), 46 (), 41-54CODEN: JOAUEP; ISSN:0896-8411. (Elsevier Ltd.)A review. Autoimmune hepatitis (AIH), primary biliary cirrhosis (PBC) and primary sclerosing cholangitis (PSC) are complex disorders, resulting from the interaction of genetic and environmental factors. For many years, investigators have attempted to delineate the genetic architecture of these conditions, aiming to elucidate disease pathogenesis and identify mol. targets for pharmacotherapy. Early genetic studies consisted of HLA assocn. studies and non-HLA candidate gene assocn. studies, designed to identify assocn. with selected HLA or non-HLA loci. HLA assocn. studies identified HLA risk loci that are now well-established. Non-HLA candidate gene studies were less fruitful because they were mostly underpowered to detect modest effects and were frequently designed to investigate one or two functional polymorphisms, meaning that gene coverage was poor. Furthermore, weak assocns. detected in one small cohort were often never validated. If replication studies were undertaken, the results were often conflicting. More recently, a series of genome-wide assocn. studies (GWAS) and related study designs have evaluated the impact of common genetic variants (frequency >5% in the general population) across the entire genome. These studies have identified several non-HLA risk loci for autoimmune liver disease. The majority of risk loci detected are similar to those of non-hepatic immune-mediated diseases, suggesting that outcomes from GWAS and related genetic studies reflect broad phenotypic themes rather than traditional clin. conditions. The specific genetic basis of these PBC and PSC assocd. inflammatory themes as detd. by GWAS is described and discussed in the context of interacting genetic and non-genetic (including environmental) factors.
- 37Bowcock, A. M. Genome-wide association studies and infectious disease Crit. Rev. Immunol. 2010, 30, 305– 309Google Scholar37https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXntValsL4%253D&md5=470818ec63f54f2c3d9653df2eeb156cGenome-wide association studies and infectious diseaseBowcock, Anne M.Critical Reviews in Immunology (2010), 30 (3), 305-309CODEN: CCRIDE; ISSN:1040-8401. (Begell House, Inc.)A review. The identification of genetic variants predisposing to complex diseases and phenotypes represent a challenge for geneticists in the early part of the 21st century. These are not simple Mendelian disorders caused by single mutations, such as cystic fibrosis or Huntington's disease, but common diseases that are usually polygenic in origin. The predisposing genes can be susceptibility factors or protective factors. One example of such a complex disease is the inflammatory skin disease psoriasis. However, another example could be protection from an infectious disease. Both of these phenotypes are due in part to the presence of low-risk variants in the host. Moreover, all of these complex phenotypes require environmental triggers as well and, in the case of infectious diseases, these are pathogens. In the case of other common diseases such as cardiovascular disease the triggers are often lifestyle-related issues such as diet or exercise. Genome-wide assocn. studies are now identifying some of these genetic susceptibility factors.
- 38Xu, C.; Aragam, N.; Li, X.; Villla, E. C.; Wang, L.; Briones, D.; Petty, L.; Posada, Y.; Arana, T. B.; Cruz, G.; Mao, C.; Camarillo, C.; Su, B. B.; Escamilla, M. A.; BCL9, Wang K. and C9orf5 are associated with negative symptoms in schizophrenia: meta-analysis of two genome-wide association studies PLoS One 2013, 8, e51674Google ScholarThere is no corresponding record for this reference.
- 39Wang, C.; Schroeder, F. A.; Hooker, J. M. Visualizing epigenetics: Current advances and advantages in HDAC PET imaging techniques Neuroscience 2014, 264, 186– 197Google ScholarThere is no corresponding record for this reference.
- 40Tang, J.; Yan, Y.; Zhao, T. C.; Gong, R.; Bayliss, G.; Yan, H.; Zhuang, S.; Class, I. HDAC activity is required for renal protection and regeneration after acute kidney injury Am. J. Physiol. Renal Physiol. 2014, 307, F303– 16Google ScholarThere is no corresponding record for this reference.
- 41Schroeder, F. A.; Lin, C. L.; Crusio, W. E.; Akbarian, S. Antidepressant-like effects of the histone deacetylase inhibitor, sodium butyrate, in the mouse Biol. Psychiatry 2007, 62, 55– 64Google Scholar41https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXmsFKrur0%253D&md5=a7337d9513d437a344bdc0374db1fac1Antidepressant-Like Effects of the Histone Deacetylase Inhibitor, Sodium Butyrate, in the MouseSchroeder, Frederick A.; Lin, Cong Lily; Crusio, Wim E.; Akbarian, SchahramBiological Psychiatry (2007), 62 (1), 55-64CODEN: BIPCBF; ISSN:0006-3223. (Elsevier Inc.)Chromatin remodeling, including changes in histone acetylation, might play a role in the pathophysiol. and treatment of depression. We investigated whether the histone deacetylase inhibitor sodium butyrate (SB) administered as single drug or in combination with the selective serotonin reuptake inhibitor (SSRI) fluoxetine exerts antidepressant-like effects in mice. Mice (C57BL/6J) received injections of SB, fluoxetine, or a combination of both drugs either acutely or chronically for a period of 28 days and were subjected to a battery of tests to measure anxiety and behavioral despair. Histone acetylation and expression of brain-derived neurotrophic factor (BDNF) were monitored in hippocampus and frontal cortex. Co-treatment with SB and fluoxetine resulted in a significant 20-40% decrease in immobility scores in the tail suspension test (TST), a measure for behavioral despair, both acutely and chronically. In contrast, decreased immobility after single drug regimens was limited either to the acute (fluoxetine) or chronic (SB) paradigm. Systemic injection of SB induced short-lasting histone hyperacetylation in hippocampus and frontal cortex. Among the four treatment paradigms that resulted in improved immobility scores in the TST, three were assocd. with a transient, at least 50% increase in BDNF transcript in frontal cortex, whereas changes in hippocampus were less consistent. The histone deacetylase inhibitor SB exerts antidepressant-like effects in the mouse. The therapeutic benefits and mol. actions of histone modifying drugs, including co-treatment with SSRIs and other newer generation antidepressant medications, warrant further exploration in exptl. models.
- 42Reid, A. E.; Hooker, J.; Shumay, E.; Logan, J.; Shea, C.; Kim, S. W.; Collins, S.; Xu, Y.; Volkow, N.; Fowler, J. S. Evaluation of 6-([(18)F]fluoroacetamido)-1-hexanoicanilide for PET imaging of histone deacetylase in the baboon brain Nucl. Med. Biol. 2009, 36, 247– 258Google Scholar42https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXjvVOrs74%253D&md5=cd26404f06bd35f0aab097edf3f42846Evaluation of 6-([18F]fluoroacetamido)-1-hexanoicanilide for PET imaging of histone deacetylase in the baboon brainReid, Alicia E.; Hooker, Jacob; Shumay, Elena; Logan, Jean; Shea, Colleen; Kim, Sung Won; Collins, Shanika; Xu, Youwen; Volkow, Nora; Fowler, Joanna S.Nuclear Medicine and Biology (2009), 36 (3), 247-258CODEN: NMBIEO; ISSN:0969-8051. (Elsevier Inc.)Introduction: Histone deacetylases (HDACs) are enzymes involved in epigenetic modifications that shift the balance toward chromatin condensation and silencing of gene expression. Here, we evaluate the utility of 6-([18F]fluoroacetamido)-1-hexanoicanilide ([18F]FAHA) for positron emission tomog. imaging of HDAC activity in the baboon brain. For this purpose, we assessed its in vivo biodistribution, sensitivity to HDAC inhibition, metabolic stability and the distribution of the putative metabolite [18F]fluoroacetate ([18F]FAC). Methods: [18F]FAHA and its metabolite [18F]FAC were prepd., and their in vivo biodistribution and pharmacokinetics were detd. in baboons. [18F]FAHA metab. and its sensitivity to HDAC inhibition using suberanilohydroxamic acid (SAHA) were assessed in arterial plasma and by in vitro incubation studies. The chem. form of F-18 in rodent brain was assessed by ex vivo studies. Distribution vols. for [18F]FAHA in the brain were derived. Results: [18F]FAHA was rapidly metabolized to [18F]FAC, and both labeled compds. entered the brain. [18F]FAHA exhibited regional differences in brain uptake and kinetics. In contrast, [18F]FAC showed little variation in regional brain uptake and kinetics. A kinetic anal. that takes into account the uptake of peripherally produced [18F]FAC indicated that SAHA inhibited binding of [18F]FAHA in the baboon brain dose-dependently. In vitro studies demonstrated SAHA-sensitive metab. of [18F]FAHA to [18F]FAC within the cell and diffusion of [18F]FAC out of the cell. All radioactivity in brain homogenate from rodents was [18F]FAC at 7 min postinjection of [18F]FAHA. Conclusion: The rapid metab. of [18F]FAHA to [18F]FAC in the periphery complicates the quant. anal. of HDAC in the brain. However, dose-dependent blocking studies with SAHA and kinetic modeling indicated that a specific interaction of [18F]FAHA in the brain was obsd. Validating the nature of this interaction as HDAC specific will require addnl. studies.
- 43Black, K. J.; Koller, J. M.; Snyder, A. Z.; Perlmutter, J. S. Atlas template images for nonhuman primate neuroimaging: baboon and macaque Methods Enzymol. 2004, 385, 91– 102Google ScholarThere is no corresponding record for this reference.
- 44Logan, J.; Fowler, J. S.; Volkow, N. D.; Wang, G. J.; Ding, Y. S.; Alexoff, D. L. Distribution volume ratios without blood sampling from graphical analysis of PET data J. Cereb. Blood Flow Metab. 1996, 16, 834– 840Google Scholar44https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADyaK28znvVKltw%253D%253D&md5=0a7ce805112626cbf4373d75bb3962a1Distribution volume ratios without blood sampling from graphical analysis of PET dataLogan J; Fowler J S; Volkow N D; Wang G J; Ding Y S; Alexoff D LJournal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism (1996), 16 (5), 834-40 ISSN:0271-678X.The distribution volume ratio (DVR), which is a linear function of receptor availability, is widely used as a model parameter in imaging studies. The DVR corresponds to the ratio of the DV of a receptor-containing region to a nonreceptor region and generally requires the measurement of an arterial input function. Here we propose a graphical method for determining the DVR that does not require blood sampling. This method uses data from a nonreceptor region with an average tissue-to-plasma efflux constant k2 to approximate the plasma integral. Data from positron emission tomography studies with [11C]raclopride (n = 20) and [11C]d-threo-methylphenidate ([11C]dMP) (n = 8) in which plasma data were taken and used to compare results from two graphical methods, one that uses plasma data and one that does not. k2 was 0.163 and 0.051 min-1 for [11C]raclopride and [11C]dMP, respectively. Results from both methods were very similar, and the average percentage difference between the methods was -0.11% for [11C]raclopride and 0.46% for [11C]dMP for DVR of basal ganglia (BG) to cerebellum (CB). Good agreement between the two methods was also achieved for DVR images created by both methods. This technique provides an alternative method of analysis not requiring blood sampling that gives equivalent results for the two ligands studied. It requires initial studies with blood sampling to determine the average kinetic constant and to test applicability. In some cases, it may be possible to neglect the k2 term if the BG/CB ratio becomes reasonably constant for a sufficiently long period of time over the course of the experiment.
- 45Bantscheff, M.; Hopf, C.; Savitski, M. M.; Dittmann, A.; Grandi, P.; Michon, A. M.; Schlegl, J.; Abraham, Y.; Becher, I.; Bergamini, G.; Boesche, M.; Delling, M.; Dumpelfeld, B.; Eberhard, D.; Huthmacher, C.; Mathieson, T.; Poeckel, D.; Reader, V.; Strunk, K.; Sweetman, G.; Kruse, U.; Neubauer, G.; Ramsden, N. G.; Drewes, G. Chemoproteomics profiling of HDAC inhibitors reveals selective targeting of HDAC complexes Nat. Biotechnol. 2011, 29, 255– 265Google Scholar45https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXps1ahug%253D%253D&md5=3a826d143fb793719a56fba3c410d51eChemoproteomics profiling of HDAC inhibitors reveals selective targeting of HDAC complexesBantscheff, Marcus; Hopf, Carsten; Savitski, Mikhail M.; Dittmann, Antje; Grandi, Paola; Michon, Anne-Marie; Schlegl, Judith; Abraham, Yann; Becher, Isabelle; Bergamini, Giovanna; Boesche, Markus; Delling, Manja; Duempelfeld, Birgit; Eberhard, Dirk; Huthmacher, Carola; Mathieson, Toby; Poeckel, Daniel; Reader, Valerie; Strunk, Katja; Sweetman, Gavain; Kruse, Ulrich; Neubauer, Gitte; Ramsden, Nigel G.; Drewes, GerardNature Biotechnology (2011), 29 (3), 255-265CODEN: NABIF9; ISSN:1087-0156. (Nature Publishing Group)The development of selective histone deacetylase (HDAC) inhibitors with anti-cancer and anti-inflammatory properties remains challenging in large part owing to the difficulty of probing the interaction of small mols. with megadalton protein complexes. A combination of affinity capture and quant. mass spectrometry revealed the selectivity with which 16 HDAC inhibitors target multiple HDAC complexes scaffolded by ELM-SANT domain subunits, including a novel mitotic deacetylase complex (MiDAC). Inhibitors clustered according to their target profiles with stronger binding of aminobenzamides to the HDAC NCoR complex than to the HDAC Sin3 complex. We identified several non-HDAC targets for hydroxamate inhibitors. HDAC inhibitors with distinct profiles have correspondingly different effects on downstream targets. We also identified the anti-inflammatory drug bufexamac as a class IIb (HDAC6, HDAC10) HDAC inhibitor. Our approach enables the discovery of novel targets and inhibitors and suggests that the selectivity of HDAC inhibitors should be evaluated in the context of HDAC complexes and not purified catalytic subunits.
Cited By
This article is cited by 79 publications.
- Ping Bai, Yan Liu, Liuyue Yang, Weihua Ding, Prasenjit Mondal, Na Sang, Gang Liu, Xiaoxia Lu, Thanh Tu Ho, Yanting Zhou, Rui Wu, Vishal C. Birar, Moses Q. Wilks, Rudolph E. Tanzi, Hening Lin, Can Zhang, Weimin Li, Shiqian Shen, Changning Wang. Development and Pharmacochemical Characterization Discover a Novel Brain-Permeable HDAC11-Selective Inhibitor with Therapeutic Potential by Regulating Neuroinflammation in Mice. Journal of Medicinal Chemistry 2023, Article ASAP.
- Nashaat Turkman, Sulan Xu, Chun-Han Huang, Christopher Eyermann, Julia Salino, Palwasha Khan. High-Contrast PET Imaging with [18F]NT160, a Class-IIa Histone Deacetylase Probe for In Vivo Imaging of Epigenetic Machinery in the Central Nervous System. Journal of Medicinal Chemistry 2023, 66
(8)
, 5611-5621. https://doi.org/10.1021/acs.jmedchem.2c02064
- Yan Liu, Zude Chen, Yanli Wang, Madelyn Rose Hallisey, Breanna Lizeth Varela, Anne Siewko, Darcy Tocci, Changning Wang, Yulong Xu. Noninvasive Positron Emission Tomography Imaging of SIRT1 in a Model of Early-Stage Alcoholic Liver Disease. Molecular Pharmaceutics 2023, 20
(4)
, 1990-1995. https://doi.org/10.1021/acs.molpharmaceut.2c00904
- Zhen Chen, Jiahui Chen, Laigao Chen, Chi-Hyeon Yoo, Jian Rong, Hualong Fu, Tuo Shao, Karen Coffman, Stefanus J. Steyn, April T. Davenport, James B. Daunais, Ahmed Haider, Lee Collier, Lee Josephson, Hsiao-Ying Wey, Lei Zhang, Steven H. Liang. Imaging Leucine-Rich Repeat Kinase 2 In Vivo with 18F-Labeled Positron Emission Tomography Ligand. Journal of Medicinal Chemistry 2023, 66
(3)
, 1712-1724. https://doi.org/10.1021/acs.jmedchem.2c00551
- Chi-Hyeon Yoo, Zhen Chen, Nisha Rani, Jiahui Chen, Jian Rong, Laigao Chen, Lei Zhang, Steven H. Liang, Hsiao-Ying Wey. Evaluation of [18F]PF-06455943 as a Potential LRRK2 PET Imaging Agent in the Brain of Nonhuman Primates. ACS Chemical Neuroscience 2023, 14
(3)
, 370-377. https://doi.org/10.1021/acschemneuro.2c00466
- Nina Reßing, Julian Schliehe-Diecks, Paris R. Watson, Melf Sönnichsen, Abigail D. Cragin, Andrea Schöler, Jing Yang, Linda Schäker-Hübner, Arndt Borkhardt, David W. Christianson, Sanil Bhatia, Finn K. Hansen. Development of Fluorinated Peptoid-Based Histone Deacetylase (HDAC) Inhibitors for Therapy-Resistant Acute Leukemia. Journal of Medicinal Chemistry 2022, 65
(22)
, 15457-15472. https://doi.org/10.1021/acs.jmedchem.2c01418
- Kuojun Zhang, Zhiyi Liu, Yiwu Yao, Yatao Qiu, Feng Li, Dong Chen, Dale J. Hamilton, Zheng Li, Sheng Jiang. Structure-Based Design of a Selective Class I Histone Deacetylase (HDAC) Near-Infrared (NIR) Probe for Epigenetic Regulation Detection in Triple-Negative Breast Cancer (TNBC). Journal of Medicinal Chemistry 2021, 64
(7)
, 4020-4033. https://doi.org/10.1021/acs.jmedchem.0c02161
- Tetsuro Tago, Jun Toyohara, Kenji Ishii. Preclinical Evaluation of an 18F-Labeled SW-100 Derivative for PET Imaging of Histone Deacetylase 6 in the Brain. ACS Chemical Neuroscience 2021, 12
(4)
, 746-755. https://doi.org/10.1021/acschemneuro.0c00774
- Sofie Celen, Johanna Rokka, Tonya M. Gilbert, Michel Koole, Isabeau Vermeulen, Kim Serdons, Frederick A. Schroeder, Florence F. Wagner, Tom Bleeser, Baileigh G. Hightower, Jiyun Hu, Dania Rahal, Hudson Beyzavi, Wim Vanduffel, Koen Van Laere, Janice E. Kranz, Jacob M. Hooker, Guy Bormans, Christopher J. Cawthorne. Translation of HDAC6 PET Imaging Using [18F]EKZ-001–cGMP Production and Measurement of HDAC6 Target Occupancy in Nonhuman Primates. ACS Chemical Neuroscience 2020, 11
(7)
, 1093-1101. https://doi.org/10.1021/acschemneuro.0c00074
- Seiya Hiranaka, Yuma Tega, Kei Higuchi, Toshiki Kurosawa, Yoshiharu Deguchi, Mayumi Arata, Akihiro Ito, Minoru Yoshida, Yasuo Nagaoka, Takaaki Sumiyoshi. Design, Synthesis, and Blood–Brain Barrier Transport Study of Pyrilamine Derivatives as Histone Deacetylase Inhibitors. ACS Medicinal Chemistry Letters 2018, 9
(9)
, 884-888. https://doi.org/10.1021/acsmedchemlett.8b00099
- Robin Bonomi, Vadim Popov, Maxwell T. Laws, David Gelovani, Anjoy Majhi, Aleksandr Shavrin, Xin Lu, Otto Muzik, Nashaat Turkman, Renshyan Liu, Thomas Mangner, Juri G. Gelovani. Molecular Imaging of Sirtuin1 Expression–Activity in Rat Brain Using Positron-Emission Tomography–Magnetic-Resonance Imaging with [18F]-2-Fluorobenzoylaminohexanoicanilide. Journal of Medicinal Chemistry 2018, 61
(16)
, 7116-7130. https://doi.org/10.1021/acs.jmedchem.8b00253
- Erika Bourguet, Katarzyna Ozdarska, Gautier Moroy, Jérôme Jeanblanc, Mickaël Naassila. Class I HDAC Inhibitors: Potential New Epigenetic Therapeutics for Alcohol Use Disorder (AUD). Journal of Medicinal Chemistry 2018, 61
(5)
, 1745-1766. https://doi.org/10.1021/acs.jmedchem.7b00115
- Martin G. Strebl, Arthur J. Campbell, Wen-Ning Zhao, Frederick A. Schroeder, Misha M. Riley, Peter S. Chindavong, Thomas M. Morin, Stephen J. Haggarty, Florence F. Wagner, Tobias Ritter, and Jacob M. Hooker . HDAC6 Brain Mapping with [18F]Bavarostat Enabled by a Ru-Mediated Deoxyfluorination. ACS Central Science 2017, 3
(9)
, 1006-1014. https://doi.org/10.1021/acscentsci.7b00274
- Iván Cheng-Sánchez, Cristina García-Ruiz, Guillermo A. Guerrero-Vásquez, and Francisco Sarabia . An Olefin Cross-Metathesis Approach to Depudecin and Stereoisomeric Analogues. The Journal of Organic Chemistry 2017, 82
(9)
, 4744-4757. https://doi.org/10.1021/acs.joc.7b00424
- Frederick A. Schroeder, Tonya M. Gilbert, Ningping Feng, Brendan D. Taillon, Nora D. Volkow, Robert B. Innis, Jacob M. Hooker, and Barbara K. Lipska . Expression of HDAC2 but Not HDAC1 Transcript Is Reduced in Dorsolateral Prefrontal Cortex of Patients with Schizophrenia. ACS Chemical Neuroscience 2017, 8
(3)
, 662-668. https://doi.org/10.1021/acschemneuro.6b00372
- Martin G. Strebl, Changning Wang, Frederick A. Schroeder, Michael S. Placzek, Hsiao-Ying Wey, Genevieve C. Van de Bittner, Ramesh Neelamegam, and Jacob M. Hooker . Development of a Fluorinated Class-I HDAC Radiotracer Reveals Key Chemical Determinants of Brain Penetrance. ACS Chemical Neuroscience 2016, 7
(5)
, 528-533. https://doi.org/10.1021/acschemneuro.5b00297
- Qingqing Meng, Zhiyi Liu, Feng Li, Jianjun Ma, He Wang, Yi Huan, and Zheng Li . An HDAC-Targeted Imaging Probe LBH589–Cy5.5 for Tumor Detection and Therapy Evaluation. Molecular Pharmaceutics 2015, 12
(7)
, 2469-2476. https://doi.org/10.1021/acs.molpharmaceut.5b00167
- Hsiao-Ying Wey, Changning Wang, Frederick A. Schroeder, Jean Logan, Julie C. Price, and Jacob M. Hooker . Kinetic Analysis and Quantification of [11C]Martinostat for in Vivo HDAC Imaging of the Brain. ACS Chemical Neuroscience 2015, 6
(5)
, 708-715. https://doi.org/10.1021/acschemneuro.5b00066
- Genevieve C. Van de Bittner, Emily L. Ricq, and Jacob M. Hooker . A Philosophy for CNS Radiotracer Design. Accounts of Chemical Research 2014, 47
(10)
, 3127-3134. https://doi.org/10.1021/ar500233s
- F. A. Schroeder, C. Wang, G. C. Van de Bittner, R. Neelamegam, W. R. Takakura, A. Karunakaran, H. Y. Wey, S. A. Reis, J. Gale, Y. L. Zhang, E. B. Holson, S. J. Haggarty, and J. M. Hooker . PET Imaging Demonstrates Histone Deacetylase Target Engagement and Clarifies Brain Penetrance of Known and Novel Small Molecule Inhibitors in Rat. ACS Chemical Neuroscience 2014, 5
(10)
, 1055-1062. https://doi.org/10.1021/cn500162j
- Yulia Aleksandrova, Aldar Munkuev, Evgenii Mozhaitsev, Evgenii Suslov, Dmitry Tsypyshev, Kirill Chaprov, Roman Begunov, Konstantin Volcho, Nariman Salakhutdinov, Margarita Neganova. Elaboration of the Effective Multi-Target Therapeutic Platform for the Treatment of Alzheimer’s Disease Based on Novel Monoterpene-Derived Hydroxamic Acids. International Journal of Molecular Sciences 2023, 24
(11)
, 9743. https://doi.org/10.3390/ijms24119743
- Yi Wang. The Role of the Gut Microbiota and Microbial Metabolites in the Pathogenesis
of Alzheimer’s Disease. CNS & Neurological Disorders - Drug Targets 2023, 22
(4)
, 577-598. https://doi.org/10.2174/1871527321666220417005115
- Priya Singh, Deepika Singh, Pooja Srivastava, Gauri Mishra, Anjani K. Tiwari. Evaluation of advanced, pathophysiologic new targets for imaging of CNS. Drug Development Research 2023, 84
(3)
, 484-513. https://doi.org/10.1002/ddr.22040
- Vladimir Shalgunov, Sara Lopes van den Broek, Ida Vang Andersen, Rocío García Vázquez, Nakul Ravi Raval, Mikael Palner, Yuki Mori, Gabriela Schäfer, Barbara Herrmann, Hannes Mikula, Natalie Beschorner, Maiken Nedergaard, Stina Syvänen, Matthias Barz, Gitte Moos Knudsen, Umberto Maria Battisti, Matthias Manfred Herth. Pretargeted imaging beyond the blood–brain barrier. RSC Medicinal Chemistry 2023, 14
(3)
, 444-453. https://doi.org/10.1039/D2MD00360K
- Liesbeth Everix, Elsie Neo Seane, Thomas Ebenhan, Ingeborg Goethals, Julie Bolcaen. Introducing HDAC-Targeting Radiopharmaceuticals for Glioblastoma Imaging and Therapy. Pharmaceuticals 2023, 16
(2)
, 227. https://doi.org/10.3390/ph16020227
- Chongzhao Ran, James R. Mansfield, Mingfeng Bai, Nerissa T. Viola, Abhishek Mahajan, E. James Delikatny. Practical Guidance for Developing Small-Molecule Optical Probes for In Vivo Imaging. Molecular Imaging and Biology 2023, 25
(1)
, 240-264. https://doi.org/10.1007/s11307-023-01800-1
- Brian J. Lopresti, Sarah K. Royse, Chester A. Mathis, Savannah A. Tollefson, Rajesh Narendran. Beyond monoamines: I. Novel targets and radiotracers for Positron emission tomography imaging in psychiatric disorders. Journal of Neurochemistry 2023, 164
(3)
, 364-400. https://doi.org/10.1111/jnc.15615
- Gaofeng Zhu, Ping Bai, Keren Wang, Jing Mi, Jing Yang, Jiaqi Hu, Yujuan Ban, Ran Xu, Rui Chen, Changning Wang, Lei Tang, Zhipei Sang. Design, synthesis, and evaluation of novel
O
-alkyl ferulamide derivatives as multifunctional ligands for treating Alzheimer’s disease. Journal of Enzyme Inhibition and Medicinal Chemistry 2022, 37
(1)
, 1375-1388. https://doi.org/10.1080/14756366.2022.2073442
- Ping Bai, Prasenjit Mondal, Frederick A. Bagdasarian, Nisha Rani, Yan Liu, Ashley Gomm, Darcy R. Tocci, Se Hoon Choi, Hsiao-Ying Wey, Rudolph E. Tanzi, Can Zhang, Changning Wang. Development of a potential PET probe for HDAC6 imaging in Alzheimer's disease. Acta Pharmaceutica Sinica B 2022, 12
(10)
, 3891-3904. https://doi.org/10.1016/j.apsb.2022.05.017
- Robert Hopewell, Dean Jolly, Qian Ying Li, Karen Ross, I‐Huang Tsai, Monica Lacatus‐Samoila, Jean‐Paul Soucy, Eliane Kobayashi, Pedro Rosa‐Neto, Gassan Massarweh. High‐yielding, automated radiosynthesis of [
11
C]martinostat using [
11
C]methyl triflate. Journal of Labelled Compounds and Radiopharmaceuticals 2022, 65
(6)
, 167-173. https://doi.org/10.1002/jlcr.3968
- George Crișan, Nastasia Sanda Moldovean-Cioroianu, Diana-Gabriela Timaru, Gabriel Andrieș, Călin Căinap, Vasile Chiș. Radiopharmaceuticals for PET and SPECT Imaging: A Literature Review over the Last Decade. International Journal of Molecular Sciences 2022, 23
(9)
, 5023. https://doi.org/10.3390/ijms23095023
- Alicia B. Pomilio, Arturo A. Vitale, Alberto J. Lazarowski. Neuroproteomics Chip-Based Mass Spectrometry and Other Techniques for Alzheimer’s
Disease Biomarkers – Update. Current Pharmaceutical Design 2022, 28
(14)
, 1124-1151. https://doi.org/10.2174/1381612828666220413094918
- Oliver Clauß, Linda Schäker-Hübner, Barbara Wenzel, Magali Toussaint, Winnie Deuther-Conrad, Daniel Gündel, Rodrigo Teodoro, Sladjana Dukić-Stefanović, Friedrich-Alexander Ludwig, Klaus Kopka, Peter Brust, Finn K. Hansen, Matthias Scheunemann. Development and Biological Evaluation of the First Highly Potent and Specific Benzamide-Based Radiotracer [18F]BA3 for Imaging of Histone Deacetylases 1 and 2 in Brain. Pharmaceuticals 2022, 15
(3)
, 324. https://doi.org/10.3390/ph15030324
- Mateusz Daśko, Beatriz de Pascual-Teresa, Irene Ortín, Ana Ramos. HDAC Inhibitors: Innovative Strategies for Their Design and Applications. Molecules 2022, 27
(3)
, 715. https://doi.org/10.3390/molecules27030715
- Nashaat Turkman, Daxing Liu, Isabella Pirola. Design, synthesis, biochemical evaluation, radiolabeling and in vivo imaging with high affinity class-IIa histone deacetylase inhibitor for molecular imaging and targeted therapy. European Journal of Medicinal Chemistry 2022, 228 , 114011. https://doi.org/10.1016/j.ejmech.2021.114011
- C.S. Bal, Dhritiman Chakraborthy. Gamma camera imaging in psychiatric disorders. 2022, 18-36. https://doi.org/10.1016/B978-0-12-822960-6.00222-2
- John S. Satterlee. Epigenomic Measurements in Brain Tissues. 2022, 3221-3261. https://doi.org/10.1007/978-3-030-88832-9_148
- Nashaat Turkman, Daxing Liu, Isabella Pirola. Novel late-stage radiosynthesis of 5-[18F]-trifluoromethyl-1,2,4-oxadiazole (TFMO) containing molecules for PET imaging. Scientific Reports 2021, 11
(1)
https://doi.org/10.1038/s41598-021-90069-x
- Alejandra Zúñiga-Muñoz, Wylly-Ramsés García-Niño, Roxana Carbó, Luis-Ángel Navarrete-López, Mabel Buelna-Chontal. The regulation of protein acetylation influences the redox homeostasis to protect the heart. Life Sciences 2021, 277 , 119599. https://doi.org/10.1016/j.lfs.2021.119599
- Yvonne E. Klingl, Donya Pakravan, Ludo Van Den Bosch. Opportunities for histone deacetylase inhibition in amyotrophic lateral sclerosis. British Journal of Pharmacology 2021, 178
(6)
, 1353-1372. https://doi.org/10.1111/bph.15217
- Michel Koole, Donatienne Van Weehaeghe, Kim Serdons, Marissa Herbots, Christopher Cawthorne, Sofie Celen, Frederick A. Schroeder, Jacob M. Hooker, Guy Bormans, Jan de Hoon, Janice E. Kranz, Koen Van Laere, Tonya M. Gilbert. Clinical validation of the novel HDAC6 radiotracer [18F]EKZ-001 in the human brain. European Journal of Nuclear Medicine and Molecular Imaging 2021, 48
(2)
, 596-611. https://doi.org/10.1007/s00259-020-04891-y
- Shariful A. Syed, Anthony S. Zannas. Epigenetics in psychotherapy. 2021, 701-709. https://doi.org/10.1016/B978-0-12-823577-5.00015-5
- Chieh-En J. Tseng, Tonya M. Gilbert, Mary C. Catanese, Baileigh G. Hightower, Amy T. Peters, Anjali J. Parmar, Minhae Kim, Changning Wang, Joshua L. Roffman, Hannah E. Brown, Roy H. Perlis, Nicole R. Zürcher, Jacob M. Hooker. In vivo human brain expression of histone deacetylases in bipolar disorder. Translational Psychiatry 2020, 10
(1)
https://doi.org/10.1038/s41398-020-00911-5
- Ming-Hsin Li, Han-Chih Chang, Chun-Fang Feng, Hung-Wen Yu, Chyng-Yann Shiue. Synthesis and Evaluation of 18F-INER-1577-3 as a Central Nervous System (CNS) Histone Deacetylase Imaging Agent. Current Medical Imaging Formerly Current Medical Imaging Reviews 2020, 16
(8)
, 978-990. https://doi.org/10.2174/1573405615666191008160809
- L. L. Donovan, J. H. Magnussen, A. Dyssegaard, S. Lehel, J. M. Hooker, G. M. Knudsen, H. D. Hansen. Imaging HDACs In Vivo: Cross-Validation of the [11C]Martinostat Radioligand in the Pig Brain. Molecular Imaging and Biology 2020, 22
(3)
, 569-577. https://doi.org/10.1007/s11307-019-01403-9
- Ana Xu, Feng He, Xiangna Zhang, Xiaoyang Li, Yingying Ran, Chao Wei, C. James Chou, Rui Zhang, Jingde Wu. Tacrine-hydroxamate derivatives as multitarget-directed ligands for the treatment of Alzheimer’s disease: Design, synthesis, and biological evaluation. Bioorganic Chemistry 2020, 98 , 103721. https://doi.org/10.1016/j.bioorg.2020.103721
- Zhipei Sang, Keren Wang, Ping Bai, Anguo Wu, Jian Shi, Wenmin Liu, Gaofeng Zhu, Yiling Wang, Yu Lan, Zude Chen, Yiyang Zhao, Zhanpin Qiao, Changning Wang, Zhenghuai Tan. Design, synthesis and biological evaluation of novel O-carbamoyl ferulamide derivatives as multi-target-directed ligands for the treatment of Alzheimer’s disease. European Journal of Medicinal Chemistry 2020, 194 , 112265. https://doi.org/10.1016/j.ejmech.2020.112265
- Yiwei Zhu, Stephanie A. Fiedler, Matthew L. Hibert, Changning Wang. Synthesis of a carbon-11 radiolabeled BACE1 inhibitor. Medicinal Chemistry Research 2020, 29
(2)
, 262-267. https://doi.org/10.1007/s00044-019-02480-9
- Stuart P. McCluskey, Christophe Plisson, Eugenii A. Rabiner, Oliver Howes. Advances in CNS PET: the state-of-the-art for new imaging targets for pathophysiology and drug development. European Journal of Nuclear Medicine and Molecular Imaging 2020, 47
(2)
, 451-489. https://doi.org/10.1007/s00259-019-04488-0
- Tetsuro Tago, Jun Toyohara, Kenji Ishii. Radiosynthesis and preliminary evaluation of an
18
F‐labeled tubastatin A analog for PET imaging of histone deacetylase 6. Journal of Labelled Compounds and Radiopharmaceuticals 2020, 63
(2)
, 85-95. https://doi.org/10.1002/jlcr.3823
- Jacob Peedicayil. Targeting histone modifications in psychotic disorders. 2020, 295-310. https://doi.org/10.1016/B978-0-12-816422-8.00012-X
- L. Donovan, J.H. Magnussen, A. Dyssegaard, S. Lehel, J.M. Hooker, G.M. Knudsen, H.D. Hansen. P.182 Imaging histone deacetylases in vivo: cross-validation of the [11C]Martinostat radiotracer in the pig brain. European Neuropsychopharmacology 2019, 29 , S139-S140. https://doi.org/10.1016/j.euroneuro.2019.09.226
- Tonya M. Gilbert, Nicole R. Zürcher, Mary C. Catanese, Chieh-En J. Tseng, Maria A. Di Biase, Amanda E. Lyall, Baileigh G. Hightower, Anjali J. Parmar, Anisha Bhanot, Christine J. Wu, Matthew L. Hibert, Minhae Kim, Umar Mahmood, Steven M. Stufflebeam, Frederick A. Schroeder, Changning Wang, Joshua L. Roffman, Daphne J. Holt, Douglas N. Greve, Ofer Pasternak, Marek Kubicki, Hsiao-Ying Wey, Jacob M. Hooker. Neuroepigenetic signatures of age and sex in the living human brain. Nature Communications 2019, 10
(1)
https://doi.org/10.1038/s41467-019-11031-0
- Yu Lan, Ping Bai, Zude Chen, Ramesh Neelamegam, Michael S. Placzek, Hao Wang, Stephanie A. Fiedler, Jing Yang, Gengyang Yuan, Xiying Qu, Hayden R. Schmidt, Jinchun Song, Marc D. Normandin, Chongzhao Ran, Changning Wang. Novel radioligands for imaging sigma-1 receptor in brain using positron emission tomography (PET). Acta Pharmaceutica Sinica B 2019, 9
(6)
, 1204-1215. https://doi.org/10.1016/j.apsb.2019.07.002
- Ping Bai, Hsiao-Ying Wey, Debasis Patnaik, Xiaoxia Lu, Yu Lan, Johanna Rokka, Fiedler Stephanie, Stephen J. Haggarty, Changning Wang. Positron emission tomography probes targeting bromodomain and extra-terminal (BET) domains to enable
in vivo
neuroepigenetic imaging. Chemical Communications 2019, 55
(86)
, 12932-12935. https://doi.org/10.1039/C9CC06734E
- Koen Vermeulen, Muneer Ahamed, Kaat Luyten, Guy Bormans. Evaluation of [11C]KB631 as a PET tracer for in vivo visualisation of HDAC6 in B16.F10 melanoma. Nuclear Medicine and Biology 2019, 74-75 , 1-11. https://doi.org/10.1016/j.nucmedbio.2019.05.004
- Erika Bourguet, Mickaël Naassila. How could histone deacetylase activators be useful leads in the search for new therapeutics?. Future Medicinal Chemistry 2019, 11
(11)
, 1241-1243. https://doi.org/10.4155/fmc-2019-0036
- L. Mahady, M. Nadeem, M. Malek‐Ahmadi, K. Chen, S. E. Perez, E. J. Mufson. HDAC
2 dysregulation in the nucleus basalis of Meynert during the progression of Alzheimer's disease. Neuropathology and Applied Neurobiology 2019, 45
(4)
, 380-397. https://doi.org/10.1111/nan.12518
- Tonya M. Gilbert, Nicole R. Zürcher, Christine J. Wu, Anisha Bhanot, Baileigh G. Hightower, Minhae Kim, Daniel S. Albrecht, Hsiao-Ying Wey, Frederick A. Schroeder, Anais Rodriguez-Thompson, Thomas M. Morin, Kamber L. Hart, Amelia M. Pellegrini, Misha M. Riley, Changning Wang, Steven M. Stufflebeam, Stephen J. Haggarty, Daphne J. Holt, Marco L. Loggia, Roy H. Perlis, Hannah E. Brown, Joshua L. Roffman, Jacob M. Hooker. PET neuroimaging reveals histone deacetylase dysregulation in schizophrenia. Journal of Clinical Investigation 2018, 129
(1)
, 364-372. https://doi.org/10.1172/JCI123743
- Yeong Jung, Yoon Kim, Mridula Bhalla, Sung Lee, Jinsoo Seo. Genomics: New Light on Alzheimer’s Disease Research. International Journal of Molecular Sciences 2018, 19
(12)
, 3771. https://doi.org/10.3390/ijms19123771
- Adriane Dallanora Henriques, Andrea Lessa Benedet, Einstein Francisco Camargos, Pedro Rosa-Neto, Otávio Toledo Nóbrega. Fluid and imaging biomarkers for Alzheimer's disease: Where we stand and where to head to. Experimental Gerontology 2018, 107 , 169-177. https://doi.org/10.1016/j.exger.2018.01.002
- Laura Mahady, Muhammad Nadeem, Michael Malek-Ahmadi, Kewei Chen, Sylvia E. Perez, Elliott J. Mufson. Frontal Cortex Epigenetic Dysregulation During the Progression of Alzheimer’s Disease. Journal of Alzheimer's Disease 2018, 62
(1)
, 115-131. https://doi.org/10.3233/JAD-171032
- Tetsuro Tago, Jun Toyohara. Advances in the Development of PET Ligands Targeting Histone Deacetylases for the Assessment of Neurodegenerative Diseases. Molecules 2018, 23
(2)
, 300. https://doi.org/10.3390/molecules23020300
- Grace S. Kim, Alicia K. Smith, Caroline M. Nievergelt, Monica Uddin. Neuroepigenetics of Post-Traumatic Stress Disorder. 2018, 227-253. https://doi.org/10.1016/bs.pmbts.2018.04.001
- Martin G. Strebl, Jane Yang, Lyle Isaacs, Jacob M. Hooker. Adamantane/Cucurbituril: A Potential Pretargeted Imaging Strategy in Immuno-PET. Molecular Imaging 2018, 17 , 153601211879983. https://doi.org/10.1177/1536012118799838
- José Ignacio Andrés, Mark Schmidt. Medicinal Chemistry strategies for PET tracer discovery. Drug Discovery Today: Technologies 2017, 25 , 11-17. https://doi.org/10.1016/j.ddtec.2017.10.002
- Emiri T. Mandeville, Cenk Ayata, Yi Zheng, Joseph B. Mandeville. Translational MR Neuroimaging of Stroke and Recovery. Translational Stroke Research 2017, 8
(1)
, 22-32. https://doi.org/10.1007/s12975-016-0497-z
- Mark Slifstein, Anissa Abi-Dargham. Recent Developments in Molecular Brain Imaging of Neuropsychiatric Disorders. Seminars in Nuclear Medicine 2017, 47
(1)
, 54-63. https://doi.org/10.1053/j.semnuclmed.2016.09.002
- Brian P. Rempel, Eric W. Price, Christopher P. Phenix. Molecular Imaging of Hydrolytic Enzymes Using PET and SPECT. Molecular Imaging 2017, 16 , 153601211771785. https://doi.org/10.1177/1536012117717852
- Emily L. Ricq, Jacob M. Hooker, Stephen J. Haggarty. Toward development of epigenetic drugs for central nervous system disorders: Modulating neuroplasticity
via
H3K4
methylation. Psychiatry and Clinical Neurosciences 2016, 70
(12)
, 536-550. https://doi.org/10.1111/pcn.12426
- Hsiao-Ying Wey, Tonya M. Gilbert, Nicole R. Zürcher, Angela She, Anisha Bhanot, Brendan D. Taillon, Fredrick A. Schroeder, Changing Wang, Stephen J. Haggarty, Jacob M. Hooker. Insights into neuroepigenetics through human histone deacetylase PET imaging. Science Translational Medicine 2016, 8
(351)
https://doi.org/10.1126/scitranslmed.aaf7551
- Shih-Pin Chen, Else A Tolner, Katharina Eikermann-Haerter. Animal models of monogenic migraine. Cephalalgia 2016, 36
(7)
, 704-721. https://doi.org/10.1177/0333102416645933
- John S. Satterlee. Epigenomic Measurements in Brain Tissues. 2016, 2857-2897. https://doi.org/10.1007/978-1-4939-3474-4_148
- Francesca Arena, Silvio Aime, Francesco Blasi. Contrast Media. 2016, 59-70. https://doi.org/10.1007/978-3-319-31614-7_5
- Corinde E. Wiers, Elizabeth Cabrera, Emily Skarda, Nora D. Volkow, Gene-Jack Wang. PET imaging for addiction medicine. 2016, 175-201. https://doi.org/10.1016/bs.pbr.2015.07.016
- Wei Liu, Charles Truillet, Robert R. Flavell, Thomas F. Brewer, Michael J. Evans, David M. Wilson, Christopher J. Chang. A reactivity-based [
18
F]FDG probe for in vivo formaldehyde imaging using positron emission tomography. Chemical Science 2016, 7
(8)
, 5503-5507. https://doi.org/10.1039/C6SC01503D
- Robin Bonomi, Uday Mukhopadhyay, Aleksandr Shavrin, Hsien-Hsien Yeh, Anjoy Majhi, Sajeewa W. Dewage, Amer Najjar, Xin Lu, G. Andrés Cisneros, William P. Tong, Mian M. Alauddin, Ren-Shuan Liu, Thomas J. Mangner, Nashaat Turkman, Juri G. Gelovani, . Novel Histone Deacetylase Class IIa Selective Substrate Radiotracers for PET Imaging of Epigenetic Regulation in the Brain. PLOS ONE 2015, 10
(8)
, e0133512. https://doi.org/10.1371/journal.pone.0133512
- Rieko Eguchi, Emi Yoshigai, Takamasa Koga, Satoru Kuhara, Kosuke Tashiro. Spatiotemporal expression of Prdm genes during Xenopus development. Cytotechnology 2015, 67
(4)
, 711-719. https://doi.org/10.1007/s10616-015-9846-0
- John S. Satterlee. Epigenomic Measurements in Brain Tissues. 2015, 1-41. https://doi.org/10.1007/978-1-4614-6434-1_148-1
Abstract
Figure 1
Figure 1. [11C]6: a translational PET imaging probe. We have developed a potent HDAC imaging agent, termed [11C]6, incorporating three key structural features to create a versatile and translational probe for visualizing HDAC expression in vivo. Intravenous injection of trace amounts of [11C]6 (nanogram scale) in baboon and imaging by PET-MR demonstrates quantifiable uptake in the brain and in diverse peripheral organs. This illustrates the potential for [11C]6 as a broadly applicable tool in evaluating HDAC density in humans.
Scheme 1
Scheme 1. Synthesis of 6, Its Radiolabeling Precursor (4) and [11C]6aScheme aReagents and conditions: (a) NaBH4, MeOH, overnight, rt, 75%; (b) formaldehyde, AcOH, NaBH4, MeOH, rt, overnight, 55%. (c) NH2OH (aq), 1M NaOH, MeOH/THF, 0 °C to rt, 4 h, 42% for 6, 40% for 5; RCY of [11C]6, 3–5% (non-decay corrected to trapped [11C]CH3I).
Figure 2
Figure 2. Kinetic modeling results with [11C]6 in baboon brain. (A) The total volume of distribution (VT) images from one representative animal show robust differences in radiotracer uptake at baseline and after blocking (0.5 mg/kg iv, 10 min pretreatment); (B) Two independent baseline-blocking studies were used to resolve quantitative VT data which show that pretreatment with unlabeled 6 (0.5 or 1.0 mg/kg) dose-dependently blocks tracer uptake in different baboon brain regions. WB: whole brain; CB: cerebellum; M1: primary motor cortex ; PU: putamen; TH: thalamus; V1: primary visual cortex ; CA: caudate; WM: white matter.
Figure 3
Figure 3. [11C]6 PET-MR imaging. Axial views of summed PET images (40–80 min) superimposed with MR images from the same baboon following injection of radiotracer (4 mCi/baboon). Images illustrate tracer uptake in organs of interest at baseline and after pretreatment with unlabeled 6 (0.5 mg/kg). Robust blocking was observed in organs of interest including: A, heart; B, spleen; C, kidneys; and D, pancreas. Time–activity curves (baseline, blue; blocking, red) demonstrate a high specific binding of [11C]6 in these peripheral organs as the percent injected tracer dose per cm3 tissue is markedly reduced by blocking.
References
ARTICLE SECTIONSThis article references 45 other publications.
- 1Wang, W. Y.; Pan, L.; Su, S. C.; Quinn, E. J.; Sasaki, M.; Jimenez, J. C.; Mackenzie, I. R.; Huang, E. J.; Tsai, L. H. Interaction of FUS and HDAC1 regulates DNA damage response and repair in neurons Nat. Neurosci. 2013, 16, 1383– 1391Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhsVertbzI&md5=9849ff952f2ebb722f188fb9eba5c740Interaction of FUS and HDAC1 regulates DNA damage response and repair in neuronsWang, Wen-Yuan; Pan, Ling; Su, Susan C.; Quinn, Emma J.; Sasaki, Megumi; Jimenez, Jessica C.; MacKenzie, Ian R. A.; Huang, Eric J.; Tsai, Li-HueiNature Neuroscience (2013), 16 (10), 1383-1391CODEN: NANEFN; ISSN:1097-6256. (Nature Publishing Group)Defects in DNA repair have been extensively linked to neurodegenerative diseases, but the exact mechanisms remain poorly understood. We found that FUS, an RNA/DNA-binding protein that has been linked to amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration, is important for the DNA damage response (DDR). The function of FUS in DDR involved a direct interaction with histone deacetylase 1 (HDAC1), and the recruitment of FUS to double-stranded break sites was important for proper DDR signaling. Notably, FUS proteins carrying familial ALS mutations were defective in DDR and DNA repair and showed a diminished interaction with HDAC1. Moreover, we obsd. increased DNA damage in human ALS patients harboring FUS mutations. Our findings suggest that an impaired DDR and DNA repair may contribute to the pathogenesis of neurodegenerative diseases linked to FUS mutations.
- 2Bardai, F. H.; Verma, P.; Smith, C.; Rawat, V.; Wang, L.; D’Mello, S. R. Disassociation of histone deacetylase-3 from normal huntingtin underlies mutant huntingtin neurotoxicity J. Neurosci. 2013, 33, 11833– 11838Google Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhtFKqu77F&md5=f5d39b1df5ef060d99bf39d9bdb28a2cDisassociation of histone deacetylase-3 from normal Huntingtin underlies mutant huntingtin neurotoxicityBardai, Farah H.; Verma, Pragya; Smith, Chad; Rawat, Varun; Wang, Lulu; D'Mello, Santosh R.Journal of Neuroscience (2013), 33 (29), 11833-11838CODEN: JNRSDS; ISSN:0270-6474. (Society for Neuroscience)Huntington's disease (HD) is caused by a polyglutamine expansion within the huntingtin (Htt) protein. Both loss of function of normal Htt and gain of a toxic function by the polyglutamine-expanded mutant Htt protein have been proposed to be responsible for HD, although the mol. mechanisms involved are unclear. We show that Htt is a neuroprotective protein in both HD-related and unrelated model systems. Neuroprotection by Htt is mediated by its sequestration of histone deacetylase-3 (HDAC3), a protein known to promote neuronal death. In contrast to the normal Htt, mutant Htt interacts poorly with HDAC3. However, expression of mutant Htt liberates HDAC3 from Htt, thus de-repressing its neurotoxic activity. Indeed, mutant Htt neurotoxicity is inhibited by the knockdown of HDAC3 and markedly reduced in HDAC3-deficient neurons. A redn. in Htt-HDAC3 interaction is also seen in neurons exposed to other apoptotic stimuli and in the striatum of R6/2 HD mice. Our results suggest that the robust interaction between Htt and HDAC3 along with the ability of mutant Htt to disrupt this assocn. while not itself interacting with HDAC3 provides an explanation for both the loss-of-function and gain-of-toxic-function mechanisms proposed for HD. Moreover, our results identify HDAC3 as an essential player in mutant Htt-induced neurodegeneration.
- 3Bardai, F. H.; Price, V.; Zaayman, M.; Wang, L.; D’Mello, S. R. Histone deacetylase-1 (HDAC1) is a molecular switch between neuronal survival and death J. Biol. Chem. 2012, 287, 35444– 35453Google ScholarThere is no corresponding record for this reference.
- 4Graff, J.; Rei, D.; Guan, J. S.; Wang, W. Y.; Seo, J.; Hennig, K. M.; Nieland, T. J.; Fass, D. M.; Kao, P. F.; Kahn, M.; Su, S. C.; Samiei, A.; Joseph, N.; Haggarty, S. J.; Delalle, I.; Tsai, L. H. An epigenetic blockade of cognitive functions in the neurodegenerating brain Nature 2012, 483, 222– 226Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC383ptlCjtw%253D%253D&md5=6e567f82c587972acbdf75fbb06d2954An epigenetic blockade of cognitive functions in the neurodegenerating brainGraff Johannes; Rei Damien; Guan Ji-Song; Wang Wen-Yuan; Seo Jinsoo; Hennig Krista M; Nieland Thomas J F; Fass Daniel M; Kao Patricia F; Kahn Martin; Su Susan C; Samiei Alireza; Joseph Nadine; Haggarty Stephen J; Delalle Ivana; Tsai Li-HueiNature (2012), 483 (7388), 222-6 ISSN:.Cognitive decline is a debilitating feature of most neurodegenerative diseases of the central nervous system, including Alzheimer's disease. The causes leading to such impairment are only poorly understood and effective treatments are slow to emerge. Here we show that cognitive capacities in the neurodegenerating brain are constrained by an epigenetic blockade of gene transcription that is potentially reversible. This blockade is mediated by histone deacetylase 2, which is increased by Alzheimer's-disease-related neurotoxic insults in vitro, in two mouse models of neurodegeneration and in patients with Alzheimer's disease. Histone deacetylase 2 associates with and reduces the histone acetylation of genes important for learning and memory, which show a concomitant decrease in expression. Importantly, reversing the build-up of histone deacetylase 2 by short-hairpin-RNA-mediated knockdown unlocks the repression of these genes, reinstates structural and synaptic plasticity, and abolishes neurodegeneration-associated memory impairments. These findings advocate for the development of selective inhibitors of histone deacetylase 2 and suggest that cognitive capacities following neurodegeneration are not entirely lost, but merely impaired by this epigenetic blockade.
- 5Zhang, L.; Sheng, S.; Qin, C. The role of HDAC6 in Alzheimer’s disease J. Alzheimer’s Dis. 2013, 33, 283– 295Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhvVeksrbJ&md5=23363ee9d90e437ce4a8a017cd05c87eThe Role of HDAC6 in Alzheimer's DiseaseZhang, Ling; Sheng, Shuli; Qin, ChuanJournal of Alzheimer's Disease (2013), 33 (2), 283-295CODEN: JADIF9; ISSN:1387-2877. (IOS Press)A review. The expression of histone deacetylase 6 (HDAC6)-a versatile enzyme with a known role in epigenetics-increases significantly in the hippocampus and other relevant brain regions in both patients with Alzheimer's disease (AD) and animal models of AD. However, when and how HDAC6 expression increases during the course of AD progression remains unclear. Whether HDAC6 overexpression is an underlying cause of AD or a condition resulting from AD is controversial. Mounting evidence suggests that increased HDAC6 expression contributes to AD-assocd. neurodegeneration, although beneficial effects have also been identified. This review article addresses recent research on HDAC6 structure and function, and highlights the potential detrimental and protective roles of HDAC6 overexpression in AD. We hope to shed light on whether HDAC6 overexpression is assocd. with AD etiopathogenesis or whether it rescues AD-assocd. neurodegeneration compensatorily. Furthermore, we discuss new evidence showing that HDAC6 may be a therapeutic target for AD.
- 6Jakovcevski, M.; Bharadwaj, R.; Straubhaar, J.; Gao, G.; Gavin, D. P.; Jakovcevski, I.; Mitchell, A. C.; Akbarian, S. Prefrontal cortical dysfunction after overexpression of histone deacetylase 1 Biol. Psychiatry 2013, 74, 696– 705Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXnt1agu7g%253D&md5=88a220ddd46e10f52aa775eda5a8c761Prefrontal Cortical Dysfunction After Overexpression of Histone Deacetylase 1Jakovcevski, Mira; Bharadwaj, Rahul; Straubhaar, Juerg; Gao, Guangping; Gavin, David P.; Jakovcevski, Igor; Mitchell, Amanda C.; Akbarian, SchahramBiological Psychiatry (2013), 74 (9), 696-705CODEN: BIPCBF; ISSN:0006-3223. (Elsevier)Background: Postmortem brain studies have shown that HDAC1-a lysine deacetylase with broad activity against histones and nonhistone proteins-is frequently expressed at increased levels in prefrontal cortex (PFC) of subjects diagnosed with schizophrenia and related disease. However, it remains unclear whether upregulated expression of Hdac1 in the PFC could affect cognition and behavior. Methods: Using adeno-assocd. virus, an Hdac1 transgene was expressed in young adult mouse PFC, followed by behavioral assays for working and long-term memory, repetitive activity, and response to novelty. Prefrontal cortex transcriptomes were profiled by microarray. Antipsychotic drug effects were explored in mice treated for 21 days with haloperidol or clozapine. Results: Hdac1 overexpression in PFC neurons and astrocytes resulted in robust impairments in working memory, increased repetitive behaviors, and abnormal locomotor response profiles in novel environments. Long-term memory remained intact. Over 300 transcripts showed subtle but significant changes in Hdac1-overexpressing PFC. Major histocompatibility complex class II (MHC II)-related transcripts, including HLA-DQA1/H2-Aa, HLA-DQB1/H2-Ab1, and HLA-DRB1/H2-Eb1, located in the chromosome 6p21.3-22.1 schizophrenia and bipolar disorder risk locus, were among the subset of genes with a more robust (>1.5-fold) down-regulation in expression. Hdac1 levels declined during the course of normal PFC development. Antipsychotic drug treatment, including the atypical clozapine, did not affect Hdac1 levels in PFC but induced expression of multiple MHC II transcripts. Conclusions: Excessive HDAC1 activity, due to developmental defects or other factors, is assocd. with behavioral alterations and dysregulated expression of MHC II and other gene transcripts in the PFC.
- 7Morris, M. J.; Mahgoub, M.; Na, E. S.; Pranav, H.; Monteggia, L. M. Loss of histone deacetylase 2 improves working memory and accelerates extinction learning J. Neurosci. 2013, 33, 6401– 6411Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhtl2jtrvJ&md5=5be28a6994f7c9170ffe8c2bd535fcabLoss of histone deacetylase 2 improves working memory and accelerates extinction learningMorris, Michael J.; Mahgoub, Melissa; Na, Elisa S.; Pranav, Heena; Monteggia, Lisa M.Journal of Neuroscience (2013), 33 (15), 6401-6411CODEN: JNRSDS; ISSN:0270-6474. (Society for Neuroscience)Histone acetylation and deacetylation can be dynamically regulated in response to environmental stimuli and play important roles in learning and memory. Pharmacol. inhibition of histone deacetylases (HDACs) improves performance in learning tasks; however, many of these classical agents are "pan-HDAC" inhibitors, and their use makes it difficult to det. the roles of specific HDACs in cognitive function. We took a genetic approach using mice lacking the class I HDACs, HDAC1 or HDAC2, in postmitotic forebrain neurons to investigate the specificity or functional redundancy of these HDACs in learning and synaptic plasticity. We show that selective knock-out of Hdac2 led to a robust acceleration of the extinction rate of conditioned fear responses and a conditioned taste aversion as well as enhanced performance in an attentional set-shifting task. Hdac2 knock-out had no impact on episodic memory or motor learning, suggesting that the effects are task-dependent, with the predominant impact of HDAC2 inhibition being an enhancement in an animal's ability to rapidly adapt its behavioral strategy as a result of changes in associative contingencies. Our results demonstrate that the loss of HDAC2 improves associative learning, with no effect in nonassociative learning tasks, suggesting a specific role for HDAC2 in particular types of learning. HDAC2 may be an intriguing target for cognitive and psychiatric disorders that are characterized by an inability to inhibit behavioral responsiveness to maladaptive or no longer relevant assocns.
- 8Cao, D. J.; Wang, Z. V.; Battiprolu, P. K.; Jiang, N.; Morales, C. R.; Kong, Y.; Rothermel, B. A.; Gillette, T. G.; Hill, J. A. Histone deacetylase (HDAC) inhibitors attenuate cardiac hypertrophy by suppressing autophagy Proc. Natl. Acad. Sci. U.S.A. 2011, 108, 4123– 4128Google ScholarThere is no corresponding record for this reference.
- 9Ferguson, B. S.; Harrison, B. C.; Jeong, M. Y.; Reid, B. G.; Wempe, M. F.; Wagner, F. F.; Holson, E. B.; McKinsey, T. A. Signal-dependent repression of DUSP5 by class I HDACs controls nuclear ERK activity and cardiomyocyte hypertrophy Proc. Natl. Acad. Sci. U.S.A. 2013, 110, 9806– 9811Google ScholarThere is no corresponding record for this reference.
- 10Barnes, P. J. Corticosteroid resistance in patients with asthma and chronic obstructive pulmonary disease J. Allergy. Clin. Immunol. 2013, 131, 636– 645Google Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhvFWmtrY%253D&md5=b9f464c23387722c42b85280cc106d07Corticosteroid resistance in patients with asthma and chronic obstructive pulmonary diseaseBarnes, Peter J.Journal of Allergy and Clinical Immunology (2013), 131 (3), 636-645CODEN: JACIBY; ISSN:0091-6749. (Elsevier)A review. Reduced responsiveness to the anti-inflammatory effects of corticosteroids is a major barrier to effective management of asthma in smokers and patients with severe asthma and in the majority of patients with chronic obstructive pulmonary disease (COPD). The mol. mechanisms leading to steroid resistance are now better understood, and this has identified new targets for therapy. In patients with severe asthma, several mol. mechanisms have been identified that might account for reduced steroid responsiveness, including reduced nuclear translocation of glucocorticoid receptor (GR) α after binding corticosteroids. This might be due to modification of the GR by means of phosphorylation as a result of activation of several kinases (p38 mitogen-activated protein kinase α, p38 mitogen-activated protein kinase γ, and c-Jun N-terminal kinase 1), which in turn might be due to reduced activity and expression of phosphatases, such as mitogen-activated protein kinase phosphatase 1 and protein phosphatase A2. Other mechanisms proposed include increased expression of GRβ, which competes with and thus inhibits activated GRα; increased secretion of macrophage migration inhibitory factor; competition with the transcription factor activator protein 1; and reduced expression of histone deacetylase (HDAC) 2. HDAC2 appears to mediate the action of steroids to switch off activated inflammatory genes, but in patients with COPD, patients with severe asthma, and smokers with asthma, HDAC2 activity and expression are reduced by oxidative stress through activation of phosphoinositide 3-kinase δ. Strategies for managing steroid resistance include alternative anti-inflammatory drugs, but a novel approach is to reverse steroid resistance by increasing HDAC2 expression, which can be achieved with theophylline and phosphoinositide 3-kinase δ inhibitors. Long-acting β2-agonists can also increase steroid responsiveness by reversing GRα phosphorylation. Identifying the mol. mechanisms of steroid resistance in asthmatic patients and patients with COPD can thus lead to more effective anti-inflammatory treatments.
- 11Brochier, C.; Dennis, G.; Rivieccio, M. A.; McLaughlin, K.; Coppola, G.; Ratan, R. R.; Langley, B. Specific acetylation of p53 by HDAC inhibition prevents DNA damage-induced apoptosis in neurons J. Neurosci. 2013, 33, 8621– 8632Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXotFant7w%253D&md5=1b5b2f3f53c0802a60ab93a9bf6bc537Specific acetylation of p53 by HDAC inhibition prevents DNA damage-induced apoptosis in neuronsBrochier, Camille; Dennis, Gretel; Rivieccio, Mark A.; McLaughlin, Kathryn; Coppola, Giovanni; Ratan, Rajiv R.; Langley, BrettJournal of Neuroscience (2013), 33 (20), 8621-8632CODEN: JNRSDS; ISSN:0270-6474. (Society for Neuroscience)Histone deacetylase (HDAC) inhibitors have been used to promote neuronal survival and ameliorate neurol. dysfunction in a host of neurodegenerative disease models. The precise mol. mechanisms whereby HDAC inhibitors prevent neuronal death are currently the focus of intensive research. Here we demonstrate that HDAC inhibition prevents DNA damage-induced neurodegeneration by modifying the acetylation pattern of the tumor suppressor p53, which decreases its DNA-binding and transcriptional activation of target genes. Specifically, we identify that acetylation at K382 and K381 prevents p53 from assocg. with the pro-apoptotic PUMA gene promoter, activating transcription, and inducing apoptosis in mouse primary cortical neurons. Paradoxically, acetylation of p53 at the same lysines in various cancer cell lines leads to the induction of PUMA expression and death. Together, our data provide a mol. understanding of the specific outcomes of HDAC inhibition and suggest that strategies aimed at enhancing p53 acetylation at K381 and K382 might be therapeutically viable for capturing the beneficial effects in the CNS, without compromising tumor suppression.
- 12Zhang, Z.; Yamashita, H.; Toyama, T.; Sugiura, H.; Omoto, Y.; Ando, Y.; Mita, K.; Hamaguchi, M.; Hayashi, S.; Iwase, H. HDAC6 expression is correlated with better survival in breast cancer Clin. Cancer Res. 2004, 10, 6962– 6968Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXpslGlur4%253D&md5=c04ab7b31de3132c37a18ed1d5ff54feHDAC6 expression is correlated with better survival in breast cancerZhang, Zhenhuan; Yamashita, Hiroko; Toyama, Tatsuya; Sugiura, Hiroshi; Omoto, Yoko; Ando, Yoshiaki; Mita, Keiko; Hamaguchi, Maho; Hayashi, Shin-ichi; Iwase, HirotakaClinical Cancer Research (2004), 10 (20), 6962-6968CODEN: CCREF4; ISSN:1078-0432. (American Association for Cancer Research)The structure and function of chromatin can be altered by modifications to histone. Histone acetylation in vivo is a dynamic reversible process governed by histone acetyltransferases (HATs) and histone deacetylases (HDACs). HDAC6 is a unique isoform among the HDACs, and a gene expression pattern study, with cDNA microarray in MCF-7 cells, showed the HDAC6 gene to be late responsive, estrogen induced, and up-regulated. This led us to hypothesize that there was a link between levels of HDAC6 expression and the metastatic potential of breast cancer and also, therefore, the prognosis of these patients. In the present study, the level of HDAC6 mRNA expression was analyzed with quant. real-time reverse transcription-PCR, in 135 female patients with invasive breast cancer. HDAC6 protein expression was also detd. by immunohistochem. An assocn. was sought between HDAC6 expression and various clinicopathol. factors. HDAC6 mRNA was expressed at significantly higher levels in breast cancer patients with small tumors measuring less than 2 cm, with low histol. grade, and in estrogen receptor α- and progesterone receptor-pos. tumors. By contrast, no relationship was found between HDAC6 mRNA expression and any of the other clinicopathol. factors, namely, age, menopausal status, and axillary lymph node involvement. Patients expressing high levels of HDAC6 mRNA and protein had a better prognosis than those expressing low levels, in terms of disease-free survival. However, multivariate anal. failed to show that HDAC6 mRNA and protein are an independent prognostic factors for disease-free survival and overall survival. Furthermore, the patients with high levels of HDAC6 mRNA tended to be more responsive to endocrine treatment than those with low levels. Specific HDAC6 staining was found in the nucleus of some normal epithelial cells and in the cytoplasm of the majority of cancer cells. Although postmenopausal patients showed higher HDAC6 protein expression, there were no relationship between protein expression and any other clinicopathol. factors. We conclude that the levels of HDAC6 mRNA expression may have potential both as a marker of endocrine responsiveness and also as a prognostic indicator in breast cancer. Addnl. investigations are warranted concerning the relationship between HDAC6 expression and response to endocrine therapy.
- 13Rajendran, P.; Kidane, A. I.; Yu, T. W.; Dashwood, W. M.; Bisson, W. H.; Lohr, C. V.; Ho, E.; Williams, D. E.; Dashwood, R. H. HDAC turnover, CtIP acetylation and dysregulated DNA damage signaling in colon cancer cells treated with sulforaphane and related dietary isothiocyanates Epigenetics 2013, 8, 612– 623Google ScholarThere is no corresponding record for this reference.
- 14Cai, Y.; Geutjes, E. J.; de Lint, K.; Roepman, P.; Bruurs, L.; Yu, L. R.; Wang, W.; van Blijswijk, J.; Mohammad, H.; de Rink, I.; Bernards, R.; Baylin, S. B. The NuRD complex cooperates with DNMTs to maintain silencing of key colorectal tumor suppressor genes Oncogene 2014, 33, 2157– 2168Google ScholarThere is no corresponding record for this reference.
- 15Kim, M. G.; Pak, J. H.; Choi, W. H.; Park, J. Y.; Nam, J. H.; Kim, J. H. The relationship between cisplatin resistance and histone deacetylase isoform overexpression in epithelial ovarian cancer cell lines J. Gynecol. Oncol. 2012, 23, 182– 189Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXmt1Sls7o%253D&md5=4086ae72007b8a38be1462349d4ab2d5The relationship between cisplatin resistance and histone deacetylase isoform overexpression in epithelial ovarian cancer cell linesKim, Min-Gyun; Pak, Jhang Ho; Choi, Won Ho; Park, Jeong-Yeol; Nam, Joo-Hyun; Kim, Jong-HyeokJournal of Gynecologic Oncology (2012), 23 (3), 182-189CODEN: JGOOAH; ISSN:2005-0380. (Korean Society of Gynecologic Oncology and Colposcopy)Objective: To investigate the relationship between cisplatin resistance and histone deacetylase (HDAC) isoform overexpression in ovarian cancer cell lines. Methods: Expression of four HDAC isoforms (HDAC 1, 2, 3, and 4) in two ovarian cancer cell lines, SKOV3 and OVCAR3, exposed to various concns. of cisplatin was examd. by western blot analyses. Cells were transfected with plasmid DNA of each HDAC. The overexpression of protein and mRNA of each HDAC was confirmed by western blot and reverse transcriptase-polymerase chain reaction analyses, resp. The cell viability of the SKOV3 and OVCAR3 cells transfected with HDAC plasmid DNA was measured using the cell counting kit-8 assay after treatment with cisplatin. Results: The 50% inhibitory concn. of the SKOV3 and OVCAR3 cells can be detd. 15-24 h after treatment with 15 μg/mL cisplatin. The expression level of acetylated histone 3 protein in SKOV3 cells increased after exposure to cisplatin. Compared with control cells at 24 h after cisplatin exposure, the viability of SKOV3 cells overexpressing HDAC 1 and 3 increased by 15% and 13% (p<0.05), resp. On the other hand, OVCAR3 cells that overexpressed HDAC 2 and 4 exhibited increased cell viability by 23% and 20% (p<0.05), resp., compared with control cells 24 h after exposure to cisplatin. Conclusion: In SKOV3 and OVCAR3 epithelial ovarian cancer cell lines, the correlation between HDAC overexpression and cisplatin resistance was confirmed. However, the specific HDAC isoform assocd. with resistance to cisplatin varied depending on the ovarian cancer cell line. These results may suggest that each HDAC isoform conveys cisplatin resistance via different mechanisms.
- 16Hayashi, A.; Horiuchi, A.; Kikuchi, N.; Hayashi, T.; Fuseya, C.; Suzuki, A.; Konishi, I.; Shiozawa, T. Type-specific roles of histone deacetylase (HDAC) overexpression in ovarian carcinoma: HDAC1 enhances cell proliferation and HDAC3 stimulates cell migration with downregulation of E-cadherin Int. J. Cancer. 2010, 127, 1332– 1346Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXpt1Ohsbo%253D&md5=35c0d48c3512ea474029d0ad80956c4dType-specific roles of histone deacetylase (HDAC) overexpression in ovarian carcinoma: HDAC1 enhances cell proliferation and HDAC3 stimulates cell migration with downregulation of E-cadherinHayashi, Akiko; Horiuchi, Akiko; Kikuchi, Norihiko; Hayashi, Takuma; Fuseya, Chiho; Suzuki, Akihisa; Konishi, Ikuo; Shiozawa, TanriInternational Journal of Cancer (2010), 127 (6), 1332-1346CODEN: IJCNAW; ISSN:0020-7136. (Wiley-Liss, Inc.)Histone acetylation/deacetylation controls chromatin activity and subsequent gene transcription. Recent studies demonstrated the activation of histone deacetylases (HDACs) in various human malignancies; however, the expression and function of HDACs in ovarian tumors are not fully understood. In this study, we examd. the immunohistochem. expression of HDAC1, HDAC2 and HDAC3 using tissues obtained from 115 cases of ovarian tumors and compared it with that of Ki-67 (a growth marker), p21, and E-cadherin and clinicopathol. parameters. In addn., we analyzed the effect of specific siRNA for HDAC1, HDAC2 and HDAC3 on the expression of cell cycle-related mols. and E-cadherin to clarify the functional difference among the 3 HDACs. The results indicated that the immunohistochem. expression of nuclear HDAC1, HDAC2 and HDAC3 proteins increased stepwise in benign, borderline and malignant tumors. The expression of HDAC1 and HDAC2 was correlated with Ki-67 expression and that of HDAC3 was inversely correlated with E-cadherin expression. Among the HDACs examd., only HDAC1 was assocd. with a poor outcome, when overexpressed. Treatment with HDAC inhibitors suppressed the proliferation of ovarian cancer cells in assocn. with apoptosis. A specific siRNA for HDAC1 significantly reduced the proliferation of ovarian carcinoma cells via downregulation of cyclin A expression, but siRNA for HDAC3 reduced the cell migration with elevated E-cadherin expression. Our results suggested that HDAC1 plays an important role in the proliferation of ovarian cancer cells, whereas HDAC3 functions in cell adhesion and migration. Therefore, specific therapeutic approaches should be considered according to the HDAC subtypes.
- 17Benes, F. M.; Lim, B.; Matzilevich, D.; Walsh, J. P.; Subburaju, S.; Minns, M. Regulation of the GABA cell phenotype in hippocampus of schizophrenics and bipolars Proc. Natl. Acad. Sci. U.S.A. 2007, 104, 10164– 10169Google ScholarThere is no corresponding record for this reference.
- 18Broide, R. S.; Redwine, J. M.; Aftahi, N.; Young, W.; Bloom, F. E.; Winrow, C. J. Distribution of histone deacetylases 1–11 in the rat brain J. Mol. Neurosci. 2007, 31, 47– 58Google Scholar18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXls1GqtL8%253D&md5=8b0ef986705015f17dee856b602a99eaDistribution of histone deacetylases 1-11 in the rat brainBroide, Ron S.; Redwine, Jeff M.; Aftahi, Najla; Young, Warren; Bloom, Floyd E.; Winrow, Christopher J.Journal of Molecular Neuroscience (2007), 31 (1), 47-58CODEN: JMNEES; ISSN:0895-8696. (Humana Press Inc.)Although protein phosphorylation has been characterized more extensively, modulation of the acetylation state of signaling mols. is now being recognized as a key means of signal transduction. The enzymes responsible for mediating these changes include histone acetyltransferases and histone deacetylases (HDACs). Members of the HDAC family of enzymes have been identified as potential therapeutic targets for diseases ranging from cancer to ischemia and neurodegeneration. The authors initiated a project to conduct comprehensive gene expression mapping of the 11 HDAC isoforms (HDAC1-11) (classes I, II, and IV) throughout the rat brain using high-resoln. in situ hybridization (ISH) and imaging technol. Internal and external data bases were employed to identify the appropriate rat sequence information for probe selection. In addn., immunohistochem. was performed on these samples to sep. examine HDAC expression in neurons, astrocytes, oligodendrocytes, and endothelial cells in the CNS. This double-labeling approach enabled the identification of specific cell types in which the individual HDACs were expressed. The signals obtained by ISH were compared to radiolabeled stds. and thereby enabled semiquant. anal. of individual HDAC isoforms and defined relative levels of gene expression in >50 brain regions. This project produced an extensive atlas of 11 HDAC isoforms throughout the rat brain, including cell type localization, providing a valuable resource for examg. the roles of specific HDACs in the brain and the development of future modulators of HDAC activity.
- 19Hooker, J. M.; Kim, S. W.; Alexoff, D.; Xu, Y.; Shea, C.; Reid, A.; Volkow, N.; Fowler, J. S. Histone deacetylase inhibitor, MS-275, exhibits poor brain penetration: PK studies of [11C]MS-275 using Positron Emission Tomography ACS Chem. Neurosci. 2010, 1, 65– 73Google Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhtlKms7vL&md5=f1cf8e753ac80029fae6cf77193f172fHistone Deacetylase Inhibitor MS-275 Exhibits Poor Brain Penetration: Pharmacokinetic Studies of [11C]MS-275 using Positron Emission TomographyHooker, Jacob M.; Kim, Sung Won; Alexoff, David; Xu, Youwen; Shea, Colleen; Reid, Alicia; Volkow, Nora; Fowler, Joanna S.ACS Chemical Neuroscience (2010), 1 (1), 65-73CODEN: ACNCDM; ISSN:1948-7193. (American Chemical Society)MS-275 (entinostat) is a histone deacetylase (HDAC) inhibitor currently in clin. trials for the treatment of several types of cancer. Recent reports have noted that MS-275 can cross the blood-brain barrier (BBB) and cause region-specific changes in rodent brain histone acetylation. To characterize the pharmacokinetics and distribution of MS-275 in the brain using positron emission tomog. (PET), we labeled the carbamate carbon of MS-275 with carbon-11. Using PET, we detd. that [11C]MS-275 has low uptake in brain tissue when administered i.v. to nonhuman primates. In rodent studies, we obsd. that pharmacokinetics and brain accumulation of [11C]MS-275 were not changed by the coadministration of large doses of unlabeled MS-275. These results, which both highlight the poor brain penetration of MS-275, clearly suggest its limitation as a therapeutic agent for the central nervous system (CNS). Moreover, our study demonstrates the effectiveness of PET at providing brain pharmacokinetic data for HDAC inhibitors. These data are important not only for the development of new compds. for peripheral cancer treatment (where CNS exclusion is often advantageous) but also for the treatment of neurol. disorders (where CNS penetration is crit.).
- 20Yeh, H. H.; Tian, M.; Hinz, R.; Young, D.; Shavrin, A.; Mukhapadhyay, U.; Flores, L. G.; Balatoni, J.; Soghomonyan, S.; Jeong, H. J.; Pal, A.; Uthamanthil, R.; Jackson, J. N.; Nishi, R.; Mizuma, H.; Onoe, H.; Kagawa, S.; Higashi, T.; Fukumitsu, N.; Alauddin, M.; Tong, W.; Herholz, K.; Gelovani, J. G. Imaging epigenetic regulation by histone deacetylases in the brain using PET/MRI with (1)(8)F-FAHA Neuroimage 2013, 64, 630– 639Google ScholarThere is no corresponding record for this reference.
- 21Hendricks, J. A.; Keliher, E. J.; Marinelli, B.; Reiner, T.; Weissleder, R.; Mazitschek, R. In vivo PET imaging of histone deacetylases by 18F-suberoylanilide hydroxamic acid (18F-SAHA) J. Med. Chem. 2011, 54, 5576– 5582Google ScholarThere is no corresponding record for this reference.
- 22Seo, Y. J.; Kang, Y.; Muench, L.; Reid, A.; Caesar, S.; Jean, L.; Fevier-Wagner, F.; Holson, E. B.; Haggarty, S. J.; Weiss, P.; King, P.; Carter, P.; Volkow, N. D.; Fowler, J. S.; Hooker, J. M.; Kim, S. W. Image-guided synthesis reveals potent blood-brain barrier permeable histone deacetylase inhibitors ACS Chem. Neurosci. 2014, 5, 588– 596Google Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXmvF2msb4%253D&md5=c59102403f07049034e1cd0a47161eb6Image-Guided Synthesis Reveals Potent Blood-Brain Barrier Permeable Histone Deacetylase InhibitorsSeo, Young Jun; Kang, Yeona; Muench, Lisa; Reid, Alicia; Caesar, Shannon; Jean, Logan; Wagner, Florence; Holson, Edward; Haggarty, Stephen J.; Weiss, Philipp; King, Payton; Carter, Pauline; Volkow, Nora D.; Fowler, Joanna S.; Hooker, Jacob M.; Kim, Sung WonACS Chemical Neuroscience (2014), 5 (7), 588-596CODEN: ACNCDM; ISSN:1948-7193. (American Chemical Society)Recent studies have revealed that several histone deacetylase (HDAC) inhibitors, which are used to study/treat brain diseases, show low blood-brain barrier (BBB) penetration. In addn. to low HDAC potency and selectivity obsd., poor brain penetrance may account for the high doses needed to achieve therapeutic efficacy. Here the authors report the development and evaluation of highly potent and blood-brain barrier permeable HDAC inhibitors for CNS applications based on an image-guided approach involving the parallel synthesis and radiolabeling of a series of compds. based on the benzamide HDAC inhibitor, MS-275 as a template. BBB penetration was optimized by rapid carbon-11 labeling and PET imaging in the baboon model and using the imaging derived data on BBB penetration from each compd. to feed back into the design process. A total of 17 compds. were evaluated, revealing mols. with both high binding affinity and BBB permeability. A key element conferring BBB penetration in this benzamide series was a basic benzylic amine. These derivs. exhibited 1-100 nM inhibitory activity against recombinant human HDAC1 and HDAC2. Three of the carbon-11 labeled aminomethyl benzamide derivs. showed high BBB penetration (∼0.015%ID/cc) and regional binding heterogeneity in the brain (high in thalamus and cerebellum). Taken together this approach has afforded a strategy and a predictive model for developing highly potent and BBB permeable HDAC inhibitors for CNS applications and for the discovery of novel candidate mols. for small mol. probes and drugs.
- 23Seo, Y. J.; Muench, L.; Reid, A.; Chen, J.; Kang, Y.; Hooker, J. M.; Volkow, N. D.; Fowler, J. S.; Kim, S. W. Radionuclide labeling and evaluation of candidate radioligands for PET imaging of histone deacetylase in the brain Bioorg. Med. Chem. Lett. 2013, 23, 6700– 6705Google ScholarThere is no corresponding record for this reference.
- 24Hanson, J. E.; La, H.; Plise, E.; Chen, Y. H.; Ding, X.; Hanania, T.; Sabath, E. V.; Alexandrov, V.; Brunner, D.; Leahy, E.; Steiner, P.; Liu, L.; Scearce-Levie, K.; Zhou, Q. SAHA enhances synaptic function and plasticity in vitro but has limited brain availability in vivo and does not impact cognition PLoS One 2013, 8, e69964Google ScholarThere is no corresponding record for this reference.
- 25Maugh, T. H., 2nd Panel urges wide use of antiviral drug Science 1979, 206, 1058– 1060Google ScholarThere is no corresponding record for this reference.
- 26Banister, S. D.; Wilkinson, S. M.; Longworth, M.; Stuart, J.; Apetz, N.; English, K.; Brooker, L.; Goebel, C.; Hibbs, D. E.; Glass, M.; Connor, M.; McGregor, I. S.; Kassiou, M. The synthesis and pharmacological evaluation of adamantane-derived indoles: cannabimimetic drugs of abuse ACS Chem. Neurosci. 2013, 4, 1081– 1092Google Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXltV2nu7c%253D&md5=859bff2f95ad6a0931b013ac532b3407The Synthesis and Pharmacological Evaluation of Adamantane-Derived Indoles: Cannabimimetic Drugs of AbuseBanister, Samuel D.; Wilkinson, Shane M.; Longworth, Mitchell; Stuart, Jordyn; Apetz, Nadine; English, Katrina; Brooker, Lance; Goebel, Catrin; Hibbs, David E.; Glass, Michelle; Connor, Mark; McGregor, Iain S.; Kassiou, MichaelACS Chemical Neuroscience (2013), 4 (7), 1081-1092CODEN: ACNCDM; ISSN:1948-7193. (American Chemical Society)Two novel adamantane derivs., adamantan-1-yl(1-pentyl-1H-indol-3-yl)methanone (AB-001) and N-(adamtan-1-yl)-1-pentyl-1H-indole-3-carboxamide (SDB-001), were recently identified as cannabimimetic indoles of abuse. Conflicting anecdotal reports of the psychoactivity of AB-001 in humans, and a complete dearth of information about the bioactivity of SDB-001, prompted the prepn. of AB-001, SDB-001, and several analogs intended to explore preliminary structure-activity relationships within this class. This study sought to elucidate which structural features of AB-001, SDB-001, and their analogs govern the cannabimimetic potency of these chemotypes in vitro and in vivo. All compds. showed similar full agonist profiles at CB1 (EC50 = 16-43 nM) and CB2 (EC50 = 29-216 nM) receptors in vitro using a FLIPR membrane potential assay, with the exception of SDB-002, which demonstrated partial agonist activity at CB2 receptors. The activity of AB-001, AB-002, and SDB-001 in rats was compared to that of Δ9-tetrahydrocannabinol (Δ9-THC) and cannabimimetic indole JWH-018 using biotelemetry. SDB-001 dose-dependently induced hypothermia and reduced heart rate (maximal dose 10 mg/kg) with potency comparable to that of Δ9-tetrahydrocannabinol (Δ9-THC, maximal dose 10 mg/kg), and lower than that of JWH-018 (maximal dose 3 mg/kg). Addnl., the changes in body temp. and heart rate affected by SDB-001 are of longer duration than those of Δ9-THC or JWH-018, suggesting a different pharmacokinetic profile. In contrast, AB-001, and its homolog, AB-002, did not produce significant hypothermic and bradycardic effects, even at relatively higher doses (up to 30 mg/kg), indicating greatly reduced potency compared to Δ9-THC, JWH-018, and SDB-001.
- 27Gopalan, B.; Ponpandian, T.; Kachhadia, V.; Bharathimohan, K.; Vignesh, R.; Sivasudar, V.; Narayanan, S.; Mandar, B.; Praveen, R.; Saranya, N.; Rajagopal, S. Discovery of adamantane based highly potent HDAC inhibitors Bioorg. Med. Chem. Lett. 2013, 23, 2532– 2537Google ScholarThere is no corresponding record for this reference.
- 28Farde, L.; Eriksson, L.; Blomquist, G.; Halldin, C. Kinetic analysis of central [11C]raclopride binding to D2-dopamine receptors studied by PET--a comparison to the equilibrium analysis J. Cereb. Blood Flow Metab. 1989, 9, 696– 708Google Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL1MXmtV2msLY%253D&md5=86b7f5f4a1d44222f2009990c889b1b7Kinetic analysis of central [11C]raclopride binding to D2-dopamine receptors studied by PET - a comparison to the equilibrium analysisFarde, L.; Eriksson, L.; Blomquist, G.; Halldin, C.Journal of Cerebral Blood Flow and Metabolism (1989), 9 (5), 696-708CODEN: JCBMDN; ISSN:0271-678X.[11C]raclopride binding to central D2-dopamine receptors in humans has previously been examd. by positron emission tomog. (PET). Based on the rapid occurrence of binding equil., a satn. anal. was developed for the detn. of receptor d. (Bmax) and affinity (Kd). For anal. of PET measurements obtained with other ligands, a kinetic 3-compartment model was used. In the present study, the brain uptake of [11C]raclopride was analyzed further by applying both a kinetic and an equil. anal. to data obtained from 4 PET expts. in each of 3 healthy subjects. First regional cerebral blood vol. was detd. In the second and third expt., [11C]raclopride with high and low specific activity was used. In a fourth expt., the [11C]raclopride enantiomer [11C]FLB472 was used to examine the concn. of free radioligand and nonspecific binding in brain. Radioactivity in arterial blood was measured using an automated blood sampling system. Bmax And Kd values for [11C]raclopride binding could be detd. also with the kinetic anal. As expected theor., those values were similar to those obtained with the equil. anal. In addn., the kinetic anal. allowed sep. detn. of the assocn. and dissocn. rate consts., kon and koff, resp. Examn. of [11C]raclopride and [11C]FLB472 uptake in brain regions devoid of specific D2-dopamine receptor binding indicated a fourth compartment in which uptake was reversible, nonstereoselective, and nonsaturable in the dose range studied.
- 29Innis, R. B.; Cunningham, V. J.; Delforge, J.; Fujita, M.; Gjedde, A.; Gunn, R. N.; Holden, J.; Houle, S.; Huang, S. C.; Ichise, M.; Iida, H.; Ito, H.; Kimura, Y.; Koeppe, R. A.; Knudsen, G. M.; Knuuti, J.; Lammertsma, A. A.; Laruelle, M.; Logan, J.; Maguire, R. P.; Mintun, M. A.; Morris, E. D.; Parsey, R.; Price, J. C.; Slifstein, M.; Sossi, V.; Suhara, T.; Votaw, J. R.; Wong, D. F.; Carson, R. E. Consensus nomenclature for in vivo imaging of reversibly binding radioligands J. Cereb. Blood Flow Metab. 2007, 27, 1533– 1539Google Scholar29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXptlSqtrc%253D&md5=77f7e875203e18e1a15d3e2d7fcd4ac6Consensus nomenclature for in vivo imaging of reversibly binding radioligandsInnis, Robert B.; Cunningham, Vincent J.; Delforge, Jacques; Fujita, Masahiro; Gjedde, Albert; Gunn, Roger N.; Holden, James; Houle, Sylvain; Huang, Sung-Cheng; Ichise, Masanori; Iida, Hidehiro; Ito, Hiroshi; Kimura, Yuichi; Koeppe, Robert A.; Knudsen, Gitte M.; Knuuti, Juhani; Lammertsma, Adriaan A.; Laruelle, Marc; Logan, Jean; Maguire, Ralph Paul; Mintun, Mark A.; Morris, Evan D.; Parsey, Ramin; Price, Julie C.; Slifstein, Mark; Sossi, Vesna; Suhara, Tetsuya; Votaw, John R.; Wong, Dean F.; Carson, Richard E.Journal of Cerebral Blood Flow & Metabolism (2007), 27 (9), 1533-1539CODEN: JCBMDN; ISSN:0271-678X. (Nature Publishing Group)A review. An international group of experts in pharmacokinetic modeling recommends a consensus nomenclature to describe in vivo mol. imaging of reversibly binding radioligands.
- 30Wang, Y.; Zhang, Y. L.; Hennig, K.; Gale, J. P.; Hong, Y.; Cha, A.; Riley, M.; Wagner, F.; Haggarty, S. J.; Holson, E.; Hooker, J. Class I HDAC imaging using [ (3)H]CI-994 autoradiography Epigenetics 2013, 8, 756– 764Google ScholarThere is no corresponding record for this reference.
- 31Schroeder, F. A.; Lewis, M. C.; Fass, D. M.; Wagner, F. F.; Zhang, Y. L.; Hennig, K. M.; Gale, J.; Zhao, W. N.; Reis, S.; Barker, D. D.; Berry-Scott, E.; Kim, S. W.; Clore, E. L.; Hooker, J. M.; Holson, E. B.; Haggarty, S. J.; Petryshen, T. L. A selective HDAC 1/2 inhibitor modulates chromatin and gene expression in brain and alters mouse behavior in two mood-related tests PLoS One 2013, 8, e71323Google ScholarThere is no corresponding record for this reference.
- 32Fass, D. M.; Schroeder, F. A.; Perlis, R. H.; Haggarty, S. J. Epigenetic mechanisms in mood disorders: Targeting neuroplasticity Neuroscience 2014, 264, 112– 130Google ScholarThere is no corresponding record for this reference.
- 33Davies, G.; Tenesa, A.; Payton, A.; Yang, J.; Harris, S. E.; Liewald, D.; Ke, X.; Le Hellard, S.; Christoforou, A.; Luciano, M.; McGhee, K.; Lopez, L.; Gow, A. J.; Corley, J.; Redmond, P.; Fox, H. C.; Haggarty, P.; Whalley, L. J.; McNeill, G.; Goddard, M. E.; Espeseth, T.; Lundervold, A. J.; Reinvang, I.; Pickles, A.; Steen, V. M.; Ollier, W.; Porteous, D. J.; Horan, M.; Starr, J. M.; Pendleton, N.; Visscher, P. M.; Deary, I. J. Genome-wide association studies establish that human intelligence is highly heritable and polygenic Mol. Psychiatry 2011, 16, 996– 1005Google Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXht1CltrbM&md5=0287370fcfc4ced4dbe3d07b982cdcf0Genome-wide association studies establish that human intelligence is highly heritable and polygenicDavies, G.; Tenesa, A.; Payton, A.; Yang, J.; Harris, S. E.; Liewald, D.; Ke, X.; Le Hellard, S.; Christoforou, A.; Luciano, M.; McGhee, K.; Lopez, L.; Gow, A. J.; Corley, J.; Redmond, P.; Fox, H. C.; Haggarty, P.; Whalley, L. J.; McNeill, G.; Goddard, M. E.; Espeseth, T.; Lundervold, A. J.; Reinvang, I.; Pickles, A.; Steen, V. M.; Ollier, W.; Porteous, D. J.; Horan, M.; Starr, J. M.; Pendleton, N.; Visscher, P. M.; Deary, I. J.Molecular Psychiatry (2011), 16 (10), 996-1005CODEN: MOPSFQ; ISSN:1359-4184. (Nature Publishing Group)General intelligence is an important human quant. trait that accounts for much of the variation in diverse cognitive abilities. Individual differences in intelligence are strongly assocd. with many important life outcomes, including educational and occupational attainments, income, health and lifespan. Data from twin and family studies are consistent with a high heritability of intelligence, but this inference has been controversial. The authors conducted a genome-wide anal. of 3511 unrelated adults with data on 549 692 single nucleotide polymorphisms (SNPs) and detailed phenotypes on cognitive traits. They est. that 40% of the variation in crystd.-type intelligence and 51% of the variation in fluid-type intelligence between individuals is accounted for by linkage disequil. between genotyped common SNP markers and unknown causal variants. These ests. provide lower bounds for the narrow-sense heritability of the traits. The authors partitioned genetic variation on individual chromosomes and found that, on av., longer chromosomes explain more variation. Finally, using just SNP data the authors predicted ∼1% of the variance of crystd. and fluid cognitive phenotypes in an independent sample (P=0.009 and 0.028, resp.). These results unequivocally confirm that a substantial proportion of individual differences in human intelligence is due to genetic variation, and are consistent with many genes of small effects underlying the additive genetic influences on intelligence. Mol. Psychiatry (2011) 16, 996-1005; doi:10.1038/mp.2011.85; published online 9 August 2011.
- 34McPherson, R. From genome-wide association studies to functional genomics: new insights into cardiovascular disease Can. J. Cardiol. 2013, 29, 23– 29Google Scholar34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC3s7otVelug%253D%253D&md5=87afdc14bea7fcb43daaa8fdde3ea0ebFrom genome-wide association studies to functional genomics: new insights into cardiovascular diseaseMcPherson RuthThe Canadian journal of cardiology (2013), 29 (1), 23-9 ISSN:.Genome-wide association studies (GWASs) for coronary artery disease (CAD) have identified more than 30 variants robustly associated with CAD risk. The majority are not associated with conventional risk factors but highlight novel pathways, including cellular proliferation. Although some risk variants are nonsynonymous coding variants resulting in an amino acid change in the encoded protein, the majority are in noncoding regions of the genome and may encompass multiple signals of variable effect. The use of genetic data for development of new therapies requires the identification of causative genetic variants and elucidation of the molecular mechanisms by which they predispose to CAD. The computational and laboratory approaches for the interpretation of GWAS data are discussed with a particular focus on noncoding variants, including the study of regulatory elements, the evaluation of nonsynonymous coding variants, and expression quantitative trait locus analysis for the integration of GWAS data with genome-wide messenger RNA expression data.
- 35Torres, J. M.; Cox, N. J.; Philipson, L. H. Genome wide association studies for diabetes: perspective on results and challenges Pediatr. Diabetes 2013, 14, 90– 96Google Scholar35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXmvFKnsrY%253D&md5=300744395e3830b6b92d359a4516da28Genome wide association studies for diabetes: perspective on results and challengesTorres, J. M.; Cox, N. J.; Philipson, L. H.Pediatric Diabetes (2013), 14 (2), 90-96CODEN: PDEIBT; ISSN:1399-5448. (Wiley-Blackwell)A review. Recent results of genome wide assocn. study (GWAS) for diabetes genes, while reaching impressive tech. milestones and implicating new findings for research, have been uniformly disappointing in terms of immediate clin. utility. The relative risk assocd. with any of the newly reported genetic loci, or even considering all of them together, is far less than simply that which can be obtained by taking a history and a phys. exam. For type 2 diabetes (T2D), GWAS have implicated novel pathways, supported previously known assocns., and highlighted the importance of the beta cell and insulin secretion. Monogenic forms of diabetes, on the other hand, continue to yield interesting insights into genes controlling human beta cell function but most cases of monogenic diabetes are simply not diagnosed. Here, we briefly review recent results related to type 1, type 2 and maturity onset diabetes of youth (MODY) diabetes and suggest that future studies emphasizing quant. traits are likely to yield even more insights.
- 36Mells, G. F.; Kaser, A.; Karlsen, T. H. Novel insights into autoimmune liver diseases provided by genome-wide association studies J. Autoimmun. 2013, 46, 41– 54Google Scholar36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXht1eksLjL&md5=bc5b8b336c89d14c3c2a1f25ba1e0217Novel insights into autoimmune liver diseases provided by genome-wide association studiesMells, George F.; Kaser, Arthur; Karlsen, Tom H.Journal of Autoimmunity (2013), 46 (), 41-54CODEN: JOAUEP; ISSN:0896-8411. (Elsevier Ltd.)A review. Autoimmune hepatitis (AIH), primary biliary cirrhosis (PBC) and primary sclerosing cholangitis (PSC) are complex disorders, resulting from the interaction of genetic and environmental factors. For many years, investigators have attempted to delineate the genetic architecture of these conditions, aiming to elucidate disease pathogenesis and identify mol. targets for pharmacotherapy. Early genetic studies consisted of HLA assocn. studies and non-HLA candidate gene assocn. studies, designed to identify assocn. with selected HLA or non-HLA loci. HLA assocn. studies identified HLA risk loci that are now well-established. Non-HLA candidate gene studies were less fruitful because they were mostly underpowered to detect modest effects and were frequently designed to investigate one or two functional polymorphisms, meaning that gene coverage was poor. Furthermore, weak assocns. detected in one small cohort were often never validated. If replication studies were undertaken, the results were often conflicting. More recently, a series of genome-wide assocn. studies (GWAS) and related study designs have evaluated the impact of common genetic variants (frequency >5% in the general population) across the entire genome. These studies have identified several non-HLA risk loci for autoimmune liver disease. The majority of risk loci detected are similar to those of non-hepatic immune-mediated diseases, suggesting that outcomes from GWAS and related genetic studies reflect broad phenotypic themes rather than traditional clin. conditions. The specific genetic basis of these PBC and PSC assocd. inflammatory themes as detd. by GWAS is described and discussed in the context of interacting genetic and non-genetic (including environmental) factors.
- 37Bowcock, A. M. Genome-wide association studies and infectious disease Crit. Rev. Immunol. 2010, 30, 305– 309Google Scholar37https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXntValsL4%253D&md5=470818ec63f54f2c3d9653df2eeb156cGenome-wide association studies and infectious diseaseBowcock, Anne M.Critical Reviews in Immunology (2010), 30 (3), 305-309CODEN: CCRIDE; ISSN:1040-8401. (Begell House, Inc.)A review. The identification of genetic variants predisposing to complex diseases and phenotypes represent a challenge for geneticists in the early part of the 21st century. These are not simple Mendelian disorders caused by single mutations, such as cystic fibrosis or Huntington's disease, but common diseases that are usually polygenic in origin. The predisposing genes can be susceptibility factors or protective factors. One example of such a complex disease is the inflammatory skin disease psoriasis. However, another example could be protection from an infectious disease. Both of these phenotypes are due in part to the presence of low-risk variants in the host. Moreover, all of these complex phenotypes require environmental triggers as well and, in the case of infectious diseases, these are pathogens. In the case of other common diseases such as cardiovascular disease the triggers are often lifestyle-related issues such as diet or exercise. Genome-wide assocn. studies are now identifying some of these genetic susceptibility factors.
- 38Xu, C.; Aragam, N.; Li, X.; Villla, E. C.; Wang, L.; Briones, D.; Petty, L.; Posada, Y.; Arana, T. B.; Cruz, G.; Mao, C.; Camarillo, C.; Su, B. B.; Escamilla, M. A.; BCL9, Wang K. and C9orf5 are associated with negative symptoms in schizophrenia: meta-analysis of two genome-wide association studies PLoS One 2013, 8, e51674Google ScholarThere is no corresponding record for this reference.
- 39Wang, C.; Schroeder, F. A.; Hooker, J. M. Visualizing epigenetics: Current advances and advantages in HDAC PET imaging techniques Neuroscience 2014, 264, 186– 197Google ScholarThere is no corresponding record for this reference.
- 40Tang, J.; Yan, Y.; Zhao, T. C.; Gong, R.; Bayliss, G.; Yan, H.; Zhuang, S.; Class, I. HDAC activity is required for renal protection and regeneration after acute kidney injury Am. J. Physiol. Renal Physiol. 2014, 307, F303– 16Google ScholarThere is no corresponding record for this reference.
- 41Schroeder, F. A.; Lin, C. L.; Crusio, W. E.; Akbarian, S. Antidepressant-like effects of the histone deacetylase inhibitor, sodium butyrate, in the mouse Biol. Psychiatry 2007, 62, 55– 64Google Scholar41https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXmsFKrur0%253D&md5=a7337d9513d437a344bdc0374db1fac1Antidepressant-Like Effects of the Histone Deacetylase Inhibitor, Sodium Butyrate, in the MouseSchroeder, Frederick A.; Lin, Cong Lily; Crusio, Wim E.; Akbarian, SchahramBiological Psychiatry (2007), 62 (1), 55-64CODEN: BIPCBF; ISSN:0006-3223. (Elsevier Inc.)Chromatin remodeling, including changes in histone acetylation, might play a role in the pathophysiol. and treatment of depression. We investigated whether the histone deacetylase inhibitor sodium butyrate (SB) administered as single drug or in combination with the selective serotonin reuptake inhibitor (SSRI) fluoxetine exerts antidepressant-like effects in mice. Mice (C57BL/6J) received injections of SB, fluoxetine, or a combination of both drugs either acutely or chronically for a period of 28 days and were subjected to a battery of tests to measure anxiety and behavioral despair. Histone acetylation and expression of brain-derived neurotrophic factor (BDNF) were monitored in hippocampus and frontal cortex. Co-treatment with SB and fluoxetine resulted in a significant 20-40% decrease in immobility scores in the tail suspension test (TST), a measure for behavioral despair, both acutely and chronically. In contrast, decreased immobility after single drug regimens was limited either to the acute (fluoxetine) or chronic (SB) paradigm. Systemic injection of SB induced short-lasting histone hyperacetylation in hippocampus and frontal cortex. Among the four treatment paradigms that resulted in improved immobility scores in the TST, three were assocd. with a transient, at least 50% increase in BDNF transcript in frontal cortex, whereas changes in hippocampus were less consistent. The histone deacetylase inhibitor SB exerts antidepressant-like effects in the mouse. The therapeutic benefits and mol. actions of histone modifying drugs, including co-treatment with SSRIs and other newer generation antidepressant medications, warrant further exploration in exptl. models.
- 42Reid, A. E.; Hooker, J.; Shumay, E.; Logan, J.; Shea, C.; Kim, S. W.; Collins, S.; Xu, Y.; Volkow, N.; Fowler, J. S. Evaluation of 6-([(18)F]fluoroacetamido)-1-hexanoicanilide for PET imaging of histone deacetylase in the baboon brain Nucl. Med. Biol. 2009, 36, 247– 258Google Scholar42https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXjvVOrs74%253D&md5=cd26404f06bd35f0aab097edf3f42846Evaluation of 6-([18F]fluoroacetamido)-1-hexanoicanilide for PET imaging of histone deacetylase in the baboon brainReid, Alicia E.; Hooker, Jacob; Shumay, Elena; Logan, Jean; Shea, Colleen; Kim, Sung Won; Collins, Shanika; Xu, Youwen; Volkow, Nora; Fowler, Joanna S.Nuclear Medicine and Biology (2009), 36 (3), 247-258CODEN: NMBIEO; ISSN:0969-8051. (Elsevier Inc.)Introduction: Histone deacetylases (HDACs) are enzymes involved in epigenetic modifications that shift the balance toward chromatin condensation and silencing of gene expression. Here, we evaluate the utility of 6-([18F]fluoroacetamido)-1-hexanoicanilide ([18F]FAHA) for positron emission tomog. imaging of HDAC activity in the baboon brain. For this purpose, we assessed its in vivo biodistribution, sensitivity to HDAC inhibition, metabolic stability and the distribution of the putative metabolite [18F]fluoroacetate ([18F]FAC). Methods: [18F]FAHA and its metabolite [18F]FAC were prepd., and their in vivo biodistribution and pharmacokinetics were detd. in baboons. [18F]FAHA metab. and its sensitivity to HDAC inhibition using suberanilohydroxamic acid (SAHA) were assessed in arterial plasma and by in vitro incubation studies. The chem. form of F-18 in rodent brain was assessed by ex vivo studies. Distribution vols. for [18F]FAHA in the brain were derived. Results: [18F]FAHA was rapidly metabolized to [18F]FAC, and both labeled compds. entered the brain. [18F]FAHA exhibited regional differences in brain uptake and kinetics. In contrast, [18F]FAC showed little variation in regional brain uptake and kinetics. A kinetic anal. that takes into account the uptake of peripherally produced [18F]FAC indicated that SAHA inhibited binding of [18F]FAHA in the baboon brain dose-dependently. In vitro studies demonstrated SAHA-sensitive metab. of [18F]FAHA to [18F]FAC within the cell and diffusion of [18F]FAC out of the cell. All radioactivity in brain homogenate from rodents was [18F]FAC at 7 min postinjection of [18F]FAHA. Conclusion: The rapid metab. of [18F]FAHA to [18F]FAC in the periphery complicates the quant. anal. of HDAC in the brain. However, dose-dependent blocking studies with SAHA and kinetic modeling indicated that a specific interaction of [18F]FAHA in the brain was obsd. Validating the nature of this interaction as HDAC specific will require addnl. studies.
- 43Black, K. J.; Koller, J. M.; Snyder, A. Z.; Perlmutter, J. S. Atlas template images for nonhuman primate neuroimaging: baboon and macaque Methods Enzymol. 2004, 385, 91– 102Google ScholarThere is no corresponding record for this reference.
- 44Logan, J.; Fowler, J. S.; Volkow, N. D.; Wang, G. J.; Ding, Y. S.; Alexoff, D. L. Distribution volume ratios without blood sampling from graphical analysis of PET data J. Cereb. Blood Flow Metab. 1996, 16, 834– 840Google Scholar44https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADyaK28znvVKltw%253D%253D&md5=0a7ce805112626cbf4373d75bb3962a1Distribution volume ratios without blood sampling from graphical analysis of PET dataLogan J; Fowler J S; Volkow N D; Wang G J; Ding Y S; Alexoff D LJournal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism (1996), 16 (5), 834-40 ISSN:0271-678X.The distribution volume ratio (DVR), which is a linear function of receptor availability, is widely used as a model parameter in imaging studies. The DVR corresponds to the ratio of the DV of a receptor-containing region to a nonreceptor region and generally requires the measurement of an arterial input function. Here we propose a graphical method for determining the DVR that does not require blood sampling. This method uses data from a nonreceptor region with an average tissue-to-plasma efflux constant k2 to approximate the plasma integral. Data from positron emission tomography studies with [11C]raclopride (n = 20) and [11C]d-threo-methylphenidate ([11C]dMP) (n = 8) in which plasma data were taken and used to compare results from two graphical methods, one that uses plasma data and one that does not. k2 was 0.163 and 0.051 min-1 for [11C]raclopride and [11C]dMP, respectively. Results from both methods were very similar, and the average percentage difference between the methods was -0.11% for [11C]raclopride and 0.46% for [11C]dMP for DVR of basal ganglia (BG) to cerebellum (CB). Good agreement between the two methods was also achieved for DVR images created by both methods. This technique provides an alternative method of analysis not requiring blood sampling that gives equivalent results for the two ligands studied. It requires initial studies with blood sampling to determine the average kinetic constant and to test applicability. In some cases, it may be possible to neglect the k2 term if the BG/CB ratio becomes reasonably constant for a sufficiently long period of time over the course of the experiment.
- 45Bantscheff, M.; Hopf, C.; Savitski, M. M.; Dittmann, A.; Grandi, P.; Michon, A. M.; Schlegl, J.; Abraham, Y.; Becher, I.; Bergamini, G.; Boesche, M.; Delling, M.; Dumpelfeld, B.; Eberhard, D.; Huthmacher, C.; Mathieson, T.; Poeckel, D.; Reader, V.; Strunk, K.; Sweetman, G.; Kruse, U.; Neubauer, G.; Ramsden, N. G.; Drewes, G. Chemoproteomics profiling of HDAC inhibitors reveals selective targeting of HDAC complexes Nat. Biotechnol. 2011, 29, 255– 265Google Scholar45https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXps1ahug%253D%253D&md5=3a826d143fb793719a56fba3c410d51eChemoproteomics profiling of HDAC inhibitors reveals selective targeting of HDAC complexesBantscheff, Marcus; Hopf, Carsten; Savitski, Mikhail M.; Dittmann, Antje; Grandi, Paola; Michon, Anne-Marie; Schlegl, Judith; Abraham, Yann; Becher, Isabelle; Bergamini, Giovanna; Boesche, Markus; Delling, Manja; Duempelfeld, Birgit; Eberhard, Dirk; Huthmacher, Carola; Mathieson, Toby; Poeckel, Daniel; Reader, Valerie; Strunk, Katja; Sweetman, Gavain; Kruse, Ulrich; Neubauer, Gitte; Ramsden, Nigel G.; Drewes, GerardNature Biotechnology (2011), 29 (3), 255-265CODEN: NABIF9; ISSN:1087-0156. (Nature Publishing Group)The development of selective histone deacetylase (HDAC) inhibitors with anti-cancer and anti-inflammatory properties remains challenging in large part owing to the difficulty of probing the interaction of small mols. with megadalton protein complexes. A combination of affinity capture and quant. mass spectrometry revealed the selectivity with which 16 HDAC inhibitors target multiple HDAC complexes scaffolded by ELM-SANT domain subunits, including a novel mitotic deacetylase complex (MiDAC). Inhibitors clustered according to their target profiles with stronger binding of aminobenzamides to the HDAC NCoR complex than to the HDAC Sin3 complex. We identified several non-HDAC targets for hydroxamate inhibitors. HDAC inhibitors with distinct profiles have correspondingly different effects on downstream targets. We also identified the anti-inflammatory drug bufexamac as a class IIb (HDAC6, HDAC10) HDAC inhibitor. Our approach enables the discovery of novel targets and inhibitors and suggests that the selectivity of HDAC inhibitors should be evaluated in the context of HDAC complexes and not purified catalytic subunits.
Supporting Information
Supporting Information
ARTICLE SECTIONSThe results of off-target binding, mPET imaging in rodents and SAHA pretreated study in NHP were shown in Table S1, Figure S1–S6. This material is available free of charge via the Internet at http://pubs.acs.org.
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.