Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
J Med Chem ; 66(18): 13205-13246, 2023 Sep 28.
Article in English | MEDLINE | ID: mdl-37712656

ABSTRACT

Huntington's disease (HD) is caused by an expanded CAG trinucleotide repeat in exon 1 of the huntingtin (HTT) gene. We report the design of a series of HTT pre-mRNA splicing modulators that lower huntingtin (HTT) protein, including the toxic mutant huntingtin (mHTT), by promoting insertion of a pseudoexon containing a premature termination codon at the exon 49-50 junction. The resulting transcript undergoes nonsense-mediated decay, leading to a reduction of HTT mRNA transcripts and protein levels. The starting benzamide core was modified to pyrazine amide and further optimized to give a potent, CNS-penetrant, and orally bioavailable HTT-splicing modulator 27. This compound reduced canonical splicing of the HTT RNA exon 49-50 and demonstrated significant HTT-lowering in both human HD stem cells and mouse BACHD models. Compound 27 is a structurally diverse HTT-splicing modulator that may help understand the mechanism of adverse effects such as peripheral neuropathy associated with branaplam.

2.
J Nucl Med ; 64(10): 1581-1587, 2023 10.
Article in English | MEDLINE | ID: mdl-37591545

ABSTRACT

Huntington disease (HD) is a neurodegenerative disorder caused by an expanded polyglutamine (CAG) trinucleotide expansion in the huntingtin (HTT) gene that encodes the mutant huntingtin protein (mHTT). Visualization and quantification of cerebral mHTT will provide a proxy for target engagement and a means to evaluate therapeutic interventions aimed at lowering mHTT in the brain. Here, we validated the novel radioligand 11C-labeled 6-(5-((5-methoxypyridin-2-yl)methoxy)benzo[d]oxazol-2-yl)-2-methylpyridazin-3(2H)-one (11C-CHDI-180R) using PET imaging to quantify cerebral mHTT aggregates in a macaque model of HD. Methods: Rhesus macaques received MRI-guided intrastriatal delivery of a mixture of AAV2 and AAV2.retro viral vectors expressing an HTT fragment bearing 85 CAG repeats (85Q, n = 5), a control HTT fragment bearing 10 CAG repeats (10Q, n = 4), or vector diluent only (phosphate-buffered saline, n = 5). Thirty months after surgery, 90-min dynamic PET/CT imaging was used to investigate 11C-CHDI-180R brain kinetics, along with serial blood sampling to measure input function and stability of the radioligand. The total volume of distribution was calculated using a 2-tissue-compartment model as well as Logan graphical analysis for regional quantification. Immunostaining for mHTT was performed to corroborate the in vivo findings. Results: 11C-CHDI-180R displayed good metabolic stability (51.4% ± 4.0% parent in plasma at 60 min after injection). Regional time-activity curves displayed rapid uptake and reversible binding, which were described by a 2-tissue-compartment model. Logan graphical analysis was associated with the 2-tissue-compartment model (r 2 = 0.96, P < 0.0001) and used to generate parametric volume of distribution maps. Compared with controls, animals administered the 85Q fragment exhibited significantly increased 11C-CHDI-180R binding in several cortical and subcortical brain regions (group effect, P < 0.0001). No difference in 11C-CHDI-180R binding was observed between buffer and 10Q animals. The presence of mHTT aggregates in the 85Q animals was confirmed histologically. Conclusion: We validated 11C-CHDI-180R as a radioligand to visualize and quantify mHTT aggregated species in a HD macaque model. These findings corroborate our previous work in rodent HD models and show that 11C-CHDI-180R is a promising tool to assess the mHTT aggregate load and the efficacy of therapeutic strategies.


Subject(s)
Huntington Disease , Animals , Huntington Disease/metabolism , Huntingtin Protein/genetics , Positron Emission Tomography Computed Tomography , Macaca mulatta/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Positron-Emission Tomography , Disease Models, Animal
3.
Sci Rep ; 13(1): 10194, 2023 06 23.
Article in English | MEDLINE | ID: mdl-37353500

ABSTRACT

Huntington's disease (HD) is a neurodegenerative disorder caused by expanded (≥ 40) glutamine-encoding CAG repeats in the huntingtin gene, which leads to dysfunction and death of predominantly striatal and cortical neurons. While the genetic profile and clinical signs and symptoms of the disease are better known, changes in the functional architecture of the brain, especially before the clinical expression becomes apparent, are not fully and consistently characterized. In this study, we sought to uncover functional changes in the brain in the heterozygous (HET) zQ175 delta-neo (DN) mouse model at 3, 6, and 10 months of age, using resting-state functional magnetic resonance imaging (RS-fMRI). This mouse model shows molecular, cellular and circuitry alterations that worsen through age. Motor function disturbances are manifested in this model at 6 and 10 months of age. Specifically, we investigated, longitudinally, changes in co-activation patterns (CAPs) that are the transient states of brain activity constituting the resting-state networks (RSNs). Most robust changes in the temporal properties of CAPs occurred at the 10-months time point; the durations of two anti-correlated CAPs, characterized by simultaneous co-activation of default-mode like network (DMLN) and co-deactivation of lateral-cortical network (LCN) and vice-versa, were reduced in the zQ175 DN HET animals compared to the wild-type mice. Changes in the spatial properties, measured in terms of activation levels of different brain regions, during CAPs were found at all three ages and became progressively more pronounced at 6-, and 10 months of age. We then assessed the cross-validated predictive power of CAP metrics to distinguish HET animals from controls. Spatial properties of CAPs performed significantly better than the chance level at all three ages with 80% classification accuracy at 6 and 10 months of age.


Subject(s)
Huntington Disease , Mice , Animals , Huntington Disease/diagnostic imaging , Huntington Disease/genetics , Huntington Disease/metabolism , Brain/metabolism , Heterozygote , Corpus Striatum/metabolism , Neurons/metabolism , Disease Models, Animal
4.
Neurobiol Dis ; 181: 106095, 2023 06 01.
Article in English | MEDLINE | ID: mdl-36963694

ABSTRACT

Huntington's disease is an autosomal, dominantly inherited neurodegenerative disease caused by an expansion of the CAG repeats in exon 1 of the huntingtin gene. Neuronal degeneration and dysfunction that precedes regional atrophy result in the impairment of striatal and cortical circuits that affect the brain's large-scale network functionality. However, the evolution of these disease-driven, large-scale connectivity alterations is still poorly understood. Here we used resting-state fMRI to investigate functional connectivity changes in a mouse model of Huntington's disease in several relevant brain networks and how they are affected at different ages that follow a disease-like phenotypic progression. Towards this, we used the heterozygous (HET) form of the zQ175DN Huntington's disease mouse model that recapitulates aspects of human disease pathology. Seed- and Region-based analyses were performed at different ages, on 3-, 6-, 10-, and 12-month-old HET and age-matched wild-type mice. Our results demonstrate decreased connectivity starting at 6 months of age, most prominently in regions such as the retrosplenial and cingulate cortices, pertaining to the default mode-like network and auditory and visual cortices, part of the associative cortical network. At 12 months, we observe a shift towards decreased connectivity in regions such as the somatosensory cortices, pertaining to the lateral cortical network, and the caudate putamen, a constituent of the subcortical network. Moreover, we assessed the impact of distinct Huntington's Disease-like pathology of the zQ175DN HET mice on age-dependent connectivity between different brain regions and networks where we demonstrate that connectivity strength follows a non-linear, inverted U-shape pattern, a well-known phenomenon of development and normal aging. Conversely, the neuropathologically driven alteration of connectivity, especially in the default mode and associative cortical networks, showed diminished age-dependent evolution of functional connectivity. These findings reveal that in this Huntington's disease model, altered connectivity starts with cortical network aberrations which precede striatal connectivity changes, that appear only at a later age. Taken together, these results suggest that the age-dependent cortical network dysfunction seen in rodents could represent a relevant pathological process in Huntington's disease progression.


Subject(s)
Huntington Disease , Neurodegenerative Diseases , Humans , Mice , Animals , Infant , Magnetic Resonance Imaging/methods , Huntington Disease/diagnostic imaging , Huntington Disease/genetics , Huntington Disease/pathology , Neurodegenerative Diseases/pathology , Brain/pathology , Brain Mapping , Disease Models, Animal
5.
J Med Chem ; 66(1): 641-656, 2023 01 12.
Article in English | MEDLINE | ID: mdl-36548390

ABSTRACT

Therapeutic interventions are being developed for Huntington's disease (HD), a hallmark of which is mutant huntingtin protein (mHTT) aggregates. Following the advancement to human testing of two [11C]-PET ligands for aggregated mHTT, attributes for further optimization were identified. We replaced the pyridazinone ring of CHDI-180 with a pyrimidine ring and minimized off-target binding using brain homogenate derived from Alzheimer's disease patients. The major in vivo metabolic pathway via aldehyde oxidase was blocked with a 2-methyl group on the pyrimidine ring. A strategically placed ring-nitrogen on the benzoxazole core ensured high free fraction in the brain without introducing efflux. Replacing a methoxy pendant with a fluoro-ethoxy group and introducing deuterium atoms suppressed oxidative defluorination and accumulation of [18F]-signal in bones. The resulting PET ligand, CHDI-650, shows a rapid brain uptake and washout profile in non-human primates and is now being advanced to human testing.


Subject(s)
Huntington Disease , Positron-Emission Tomography , Animals , Humans , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Ligands , Positron-Emission Tomography/methods , Huntington Disease/diagnostic imaging , Huntington Disease/drug therapy , Brain/diagnostic imaging , Brain/metabolism
6.
Eur J Nucl Med Mol Imaging ; 50(1): 48-60, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36001116

ABSTRACT

PURPOSE: Huntington's disease is caused by a trinucleotide expansion in the HTT gene, which leads to aggregation of mutant huntingtin (mHTT) protein in the brain and neurotoxicity. Direct in vivo measurement of mHTT aggregates in human brain parenchyma is not yet possible. In this first-in-human study, we investigated biodistribution and dosimetry in healthy volunteers of [11C]CHDI-00485180-R ([11C]CHDI-180R) and [11C]CHDI-00485626 ([11C]CHDI-626), two tracers designed for PET imaging of aggregated mHTT in the brain that have been validated in preclinical models. METHODS: Biodistribution and radiation dosimetry studies were performed in 3 healthy volunteers (age 25.7 ± 0.5 years; 2 F) for [11C]CHDI-180R and in 3 healthy volunteers (age 35.3 ± 6.8 years; 2 F) for [11C]CHDI-626 using sequential whole-body PET-CT. Source organs were delineated in 3D using combined PET and CT data. Individual organ doses and effective doses were determined using OLINDA 2.1. RESULTS: There were no clinically relevant adverse events. The mean effective dose (ED) for [11C]CHDI-180R was 4.58 ± 0.65 µSv/MBq, with highest absorbed doses for liver (16.9 µGy/MBq), heart wall (15.9 µGy/MBq) and small intestine (15.8 µGy/MBq). Mean ED for [11C]CHDI-626 was 5.09 ± 0.06 µSv/MBq with the highest absorbed doses for the gallbladder (26.5 µGy/MBq), small intestine (20.4 µGy/MBq) and liver (19.6 µGy/MBq). Decay-corrected brain uptake curves showed promising kinetics for [11C]CHDI-180R, but for [11C]CHDI-626 an increasing signal over time was found, probably due to accumulation of a brain-penetrant metabolite. CONCLUSION: [11C]CHDI-180R and [11C]CHDI-626 are safe for in vivo PET imaging in humans. The estimated radiation burden is in line with most 11C-ligands. While [11C]CHDI-180R has promising kinetic properties in the brain, [11C]CHDI-626 is not suitable for human in vivo mHTT PET due to the possibility of a radiometabolite accumulating in brain parenchyma. TRIAL REGISTRATION: EudraCT number 2020-002129-27. CLINICALTRIALS: gov NCT05224115 (retrospectively registered).


Subject(s)
Positron Emission Tomography Computed Tomography , Radiometry , Humans , Adult , Healthy Volunteers , Tissue Distribution , Positron-Emission Tomography/methods
7.
J Cereb Blood Flow Metab ; 42(10): 1867-1878, 2022 10.
Article in English | MEDLINE | ID: mdl-35570828

ABSTRACT

Alterations in synaptic vesicle glycoprotein 2 A (SV2A) have been associated with several neuropsychiatric and neurodegenerative disorders. Therefore, SV2A positron emission tomography (PET) imaging may provide a unique tool to investigate synaptic density dynamics during disease progression and after therapeutic intervention. This study aims to extensively characterize the novel radioligand [18F]SynVesT-1 for preclinical applications. In C57Bl/6J mice (n = 39), we assessed the plasma profile of [18F]SynVesT-1, validated the use of a noninvasive image-derived input function (IDIF) compared to an arterial input function (AIF), performed a blocking study with levetiracetam (50 and 200 mg/kg, i.p.) to verify the specificity towards SV2A, examined kinetic models for volume of distribution (VT) quantification, and explored test-retest reproducibility of [18F]SynVesT-1 in the central nervous system (CNS). Plasma availability of [18F]SynVesT-1 decreased rapidly (13.4 ± 1.5% at 30 min post-injection). VT based on AIF and IDIF showed excellent agreement (r2 = 0.95, p < 0.0001) and could be reliably estimated with a 60-min acquisition. The blocking study resulted in a complete blockade with no suitable reference region. Test-retest analysis indicated good reproducibility (mean absolute variability <10%). In conclusion, [18F]SynVesT-1 is selective for SV2A with optimal kinetics representing a candidate tool to quantify CNS synaptic density non-invasively.


Subject(s)
Brain , Synaptic Vesicles , Animals , Brain/metabolism , Glycoproteins/metabolism , Levetiracetam , Mice , Positron-Emission Tomography/methods , Radiopharmaceuticals/metabolism , Reproducibility of Results , Synaptic Vesicles/metabolism
8.
Sci Transl Med ; 14(630): eabm3682, 2022 02 02.
Article in English | MEDLINE | ID: mdl-35108063

ABSTRACT

Huntington's disease (HD) is a dominantly inherited neurodegenerative disorder caused by a CAG trinucleotide expansion in the huntingtin (HTT) gene that encodes the pathologic mutant HTT (mHTT) protein with an expanded polyglutamine (polyQ) tract. Whereas several therapeutic programs targeting mHTT expression have advanced to clinical evaluation, methods to visualize mHTT protein species in the living brain are lacking. Here, we demonstrate the development and characterization of a positron emission tomography (PET) imaging radioligand with high affinity and selectivity for mHTT aggregates. This small molecule radiolabeled with 11C ([11C]CHDI-180R) allowed noninvasive monitoring of mHTT pathology in the brain and could track region- and time-dependent suppression of mHTT in response to therapeutic interventions targeting mHTT expression in a rodent model. We further showed that in these animals, therapeutic agents that lowered mHTT in the striatum had a functional restorative effect that could be measured by preservation of striatal imaging markers, enabling a translational path to assess the functional effect of mHTT lowering.


Subject(s)
Huntington Disease , Neurodegenerative Diseases , Animals , Corpus Striatum/diagnostic imaging , Corpus Striatum/metabolism , Disease Models, Animal , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Huntington Disease/diagnostic imaging , Huntington Disease/genetics , Huntington Disease/metabolism , Ligands , Neurodegenerative Diseases/pathology
9.
Exp Ther Med ; 23(2): 115, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34970338

ABSTRACT

In-stent restenosis (ISR) can pose serious challenges for cardiologists following coronary stent implantation. Early identification of patients at high risk of ISR is considered to be effective for its prevention. However, factors that can reliably predict the risk of ISR remain elusive at present. The present study aimed to investigate the possible association between plasma long non-coding RNA (lncRNA) levels and ISR. A total of 410 patients with single-vessel lesion who received drug-eluting stents (DES) were included in the present study. After 12-36 months of follow-up, coronary angiography was performed and ISR was defined as >50% diameter stenosis at follow-up. RT-qPCR was used to measure lncRNA expression. Expression of the lncRNA RNA antisense non-coding RNA at the INK4 locus (ANRIL) was found to be upregulated whereas the lncRNA homeobox A11 antisense (HOXA11-AS) was downregulated in the plasma of patients with ISR compared with that from patients without ISR (P<0.001). Logistic regression analysis revealed that ANRIL [odds ratio (OR)=2.95; 95% confidence interval (CI)=1.68-8.08] was an independent risk factor for ISR, whilst HOXA11-AS (OR=0.58; 95% CI=0.48-0.71) was found to be an independent protective factor for ISR. Receiver operating characteristic (ROC) analysis demonstrated that high ANRIL expression [area under the ROC (auROC)=0.755; 95% CI=0.702-0.803] and low HOXA11-AS levels (auROC=0.712; 95% CI=0.657-0.763) predicted a high risk for ISR, and the combined score of ANRIL and HOXA11-AS (auROC=0.844; 95% CI=0.798-0.884) was more efficient at predicting ISR than either ANRIL or HOXA11-AS alone (P<0.001). In conclusion, increased ANRIL and decreased HOXA11-AS expressions were associated with ISR. However, combined ANRIL and HOXA11-AS plasma levels proved to be more effective at predicting ISR compared with either ANRIL or HOXA11-AS alone, suggesting that the multiplex detection of lncRNAs could be used to predict ISR in the future.

10.
J Nucl Med ; 63(6): 942-947, 2022 06.
Article in English | MEDLINE | ID: mdl-34531262

ABSTRACT

Synaptic dysfunction is a primary mechanism underlying Huntington disease (HD) progression. This study investigated changes in synaptic vesicle glycoprotein 2A (SV2A) density by means of 11C-UCB-J small-animal PET imaging in the central nervous system of mice with HD. Methods: Dynamic 11C-UCB-J small-animal PET imaging was performed at clinically relevant disease stages (at 3, 7, 10, and 16 mo) in the heterozygous knock-in Q175DN mouse model of HD and wild-type littermates (16-18 mice per genotype and time point). Cerebral 11C-UCB-J analyses were performed to assess genotypic differences during presymptomatic (3 mo) and symptomatic (7-16 mo) disease stages. 11C-UCB-J binding in the spinal cord was quantified at 16 mo. 3H-UCB-J autoradiography and SV2A immunofluorescence were performed postmortem in mouse and human brain tissues. Results:11C-UCB-J binding was lower in symptomatic heterozygous mice than in wild-type littermates in parallel with disease progression (7 and 10 mo: P < 0.01; 16 mo: P < 0.0001). Specific 11C-UCB-J binding was detectable in the spinal cord, with symptomatic heterozygous mice displaying a significant reduction (P < 0.0001). 3H-UCB-J autoradiography and SV2A immunofluorescence corroborated the in vivo measurements demonstrating lower SV2A in heterozygous mice (P < 0.05). Finally, preliminary analysis of SV2A in the human brain postmortem suggested lower SV2A in HD gene carriers than in controls without dementia. Conclusion:11C-UCB-J PET detected SV2A deficits during symptomatic disease in heterozygous mice in both the brain and the spinal cord and therefore may be suitable as a novel marker of synaptic integrity widely distributed in the central nervous system. On clinical application, 11C-UCB-J PET imaging may have promise for SV2A measurement in patients with HD during disease progression and after disease-modifying therapeutic strategies.


Subject(s)
Huntington Disease , Animals , Brain/diagnostic imaging , Brain/metabolism , Disease Progression , Humans , Huntington Disease/diagnostic imaging , Huntington Disease/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Positron-Emission Tomography/methods , Pyridines/metabolism , Synaptic Vesicles/metabolism
11.
Eur J Nucl Med Mol Imaging ; 49(4): 1166-1175, 2022 03.
Article in English | MEDLINE | ID: mdl-34651228

ABSTRACT

PURPOSE: As several therapies aimed at lowering mutant huntingtin (mHTT) brain levels in Huntington's disease (HD) are currently being investigated, noninvasive positron emission tomography (PET) imaging of mHTT could be utilized to directly evaluate therapeutic efficacy and monitor disease progression. Here we characterized and longitudinally assessed the novel radioligand [11C]CHDI-626 for mHTT PET imaging in the zQ175DN mouse model of HD. METHODS: After evaluating radiometabolites and radioligand kinetics, we conducted longitudinal dynamic PET imaging at 3, 6, 9, and 13 months of age (M) in wild-type (WT, n = 17) and heterozygous (HET, n = 23) zQ175DN mice. Statistical analysis was performed to evaluate temporal and genotypic differences. Cross-sectional cohorts at each longitudinal time point were included for post-mortem [3H]CHDI-626 autoradiography. RESULTS: Despite fast metabolism and kinetics, the radioligand was suitable for PET imaging of mHTT. Longitudinal quantification could discriminate between genotypes already at premanifest stage (3 M), showing an age-associated increase in signal in HET mice in parallel with mHTT aggregate load progression, as supported by the post-mortem [3H]CHDI-626 autoradiography. CONCLUSION: With clinical evaluation underway, [11C]CHDI-626 PET imaging appears to be a suitable preclinical candidate marker to monitor natural HD progression and for the evaluation of mHTT-lowering therapies.


Subject(s)
Huntington Disease , Animals , Carbon Radioisotopes , Cross-Sectional Studies , Disease Models, Animal , Humans , Huntington Disease/metabolism , Mice , Positron-Emission Tomography/methods
12.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 52(6): 960-965, 2021 Nov.
Article in Chinese | MEDLINE | ID: mdl-34841762

ABSTRACT

OBJECTIVE: To explore the effect of nuclear factor-erythroid 2-related factor (Nrf2) pathway activation on hippocampal neuron damage in neonatal rats with bilirubin encephalopathy. METHODS: Neonatal rats were randomly assigned to a control group (Control), a model group (Model) and an Nrf2 activator TBHQ (tert-Butylhydroquinone) group (TBHQ), with 20 rats in each group. Bilirubin solution was injected through the cerebellomedullary cistern to establish the neonatal rat model of bilirubin encephalopathy. Neurobehavioral changes were observed in rats and the water content of the brain tissue was measured. Nissl staining was done to observe the damage of hippocampal neurons. TUNEL staining was used to observe the apoptosis of hippocampal neurons. Colorimetric analysis was done to determine the Caspase-3 activity in the hippocampus. The content of malondialdehyde (MDA) and reduced glutathione (GSH) and the activity of superoxide dismutase (SOD) in the hippocampus were examined by chemical analysis. qRT-PCR and Western blot were done to measure the expression of Nrf2 and heme oxygenase-l (HO-1) mRNA and proteins in the hippocampus. RESULTS: After injection of bilirubin into the cerebellomedullary cistern, the young rats in the Model group and the TBHQ group showed different degrees of neurological abnormalities, while those in the control group showed no significant neurobehavioral abnormalities. Compared with the Control group, the Model group had severe neuronal damage, and the water content of brain tissue, the apoptosis of hippocampal neurons, the activity of Caspase-3 and the content of MDA content significantly increased ( P<0.01), while the SOD activity, GSH content, the expression of Nrf2 and HO-1 mRNA and proteins significantly decreased ( P<0.05). Compared with the Model group, neuronal damage was improved in the TBHQ group, and the water content of brain tissue, apoptosis of hippocampal neurons, activity of Caspase-3 and MDA content were all significantly reduced ( P<0.01), while SOD activity, GSH content and the expression of Nrf2 and HO-1 mRNA and proteins were significantly increased ( P<0.05). CONCLUSION: Activation of the Nrf2 pathway can improve hippocampal neuronal damage in neonatal rats with bilirubin encephalopathy and inhibit neuronal apoptosis and the oxidation reaction.


Subject(s)
Kernicterus , NF-E2-Related Factor 2 , Animals , Animals, Newborn , Hippocampus , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Neurons/metabolism , Oxidative Stress , Rats , Signal Transduction
13.
Sci Rep ; 11(1): 17977, 2021 09 09.
Article in English | MEDLINE | ID: mdl-34504195

ABSTRACT

Huntington's disease (HD) is caused by a CAG trinucleotide repeat expansion in the first exon of the huntingtin (HTT) gene coding for the huntingtin (HTT) protein. The misfolding and consequential aggregation of CAG-expanded mutant HTT (mHTT) underpin HD pathology. Our interest in the life cycle of HTT led us to consider the development of high-affinity small-molecule binders of HTT oligomerized/amyloid-containing species that could serve as either cellular and in vivo imaging tools or potential therapeutic agents. We recently reported the development of PET tracers CHDI-180 and CHDI-626 as suitable for imaging mHTT aggregates, and here we present an in-depth pharmacological investigation of their binding characteristics. We have implemented an array of in vitro and ex vivo radiometric binding assays using recombinant HTT, brain homogenate-derived HTT aggregates, and brain sections from mouse HD models and humans post-mortem to investigate binding affinities and selectivity against other pathological proteins from indications such as Alzheimer's disease and spinocerebellar ataxia 1. Radioligand binding assays and autoradiography studies using brain homogenates and tissue sections from HD mouse models showed that CHDI-180 and CHDI-626 specifically bind mHTT aggregates that accumulate with age and disease progression. Finally, we characterized CHDI-180 and CHDI-626 regarding their off-target selectivity and binding affinity to beta amyloid plaques in brain sections and homogenates from Alzheimer's disease patients.


Subject(s)
Huntingtin Protein/metabolism , Huntington Disease/metabolism , Positron-Emission Tomography/methods , Protein Aggregates/genetics , Protein Aggregation, Pathological/diagnostic imaging , Radiopharmaceuticals/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Autoradiography/methods , Brain/metabolism , Disease Models, Animal , Humans , Huntingtin Protein/genetics , Huntington Disease/pathology , Immunohistochemistry/methods , Mice , Mice, Transgenic , Nitrogen Radioisotopes/metabolism , Radioactive Tracers , Radioligand Assay/methods , Recombinant Proteins/metabolism
14.
J Med Chem ; 64(16): 12003-12021, 2021 08 26.
Article in English | MEDLINE | ID: mdl-34351166

ABSTRACT

The expanded polyglutamine-containing mutant huntingtin (mHTT) protein is implicated in neuronal degeneration of medium spiny neurons in Huntington's disease (HD) for which multiple therapeutic approaches are currently being evaluated to eliminate or reduce mHTT. Development of effective and orthogonal biomarkers will ensure accurate assessment of the safety and efficacy of pharmacologic interventions. We have identified and optimized a class of ligands that bind to oligomerized/aggregated mHTT, which is a hallmark in the HD postmortem brain. These ligands are potentially useful imaging biomarkers for HD therapeutic development in both preclinical and clinical settings. We describe here the optimization of the benzo[4,5]imidazo[1,2-a]pyrimidine series that show selective binding to mHTT aggregates over Aß- and/or tau-aggregates associated with Alzheimer's disease pathology. Compound [11C]-2 was selected as a clinical candidate based on its high free fraction in the brain, specific binding in the HD mouse model, and rapid brain uptake/washout in nonhuman primate positron emission tomography imaging studies.


Subject(s)
Brain/diagnostic imaging , Heterocyclic Compounds, 3-Ring/chemistry , Huntingtin Protein/metabolism , Protein Aggregates/physiology , Pyridines/chemistry , Radiopharmaceuticals/chemistry , Alzheimer Disease , Animals , Biomarkers/metabolism , Brain/metabolism , Carbon Radioisotopes/chemistry , Female , Heterocyclic Compounds, 3-Ring/chemical synthesis , Heterocyclic Compounds, 3-Ring/pharmacokinetics , Humans , Macaca fascicularis , Male , Mice, Inbred C57BL , Molecular Structure , Positron-Emission Tomography , Pyridines/chemical synthesis , Pyridines/pharmacokinetics , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/pharmacokinetics , Rats, Sprague-Dawley , Structure-Activity Relationship
15.
Mol Imaging Biol ; 23(2): 208-219, 2021 04.
Article in English | MEDLINE | ID: mdl-33179158

ABSTRACT

PURPOSE: Our aim in this study was to compare different non-invasive pharmacokinetic models and assess test-retest reproducibility of the radioligand [11C]SCH23390 for the quantification of dopamine D1-like receptor (D1R) in both wild-type (WT) mice and heterozygous (HET) Q175DN mice as Huntington's disease (HD) model. PROCEDURES: Adult WT (n = 9) and HET (n = 14) mice underwent a 90-min [11C]SCH23390 positron emission tomography (PET) scan followed by computed tomography (CT) to evaluate the pharmacokinetic modelling in healthy and diseased conditions. Additionally, 5 WT mice and 7 HET animals received a second [11C]SCH23390 PET scan for test-retest reproducibility. Parallel assessment of the simplified reference tissue model (SRTM), the multilinear reference tissue model (MRTM) and the Logan reference tissue model (Logan Ref) using the striatum as a receptor-rich region and the cerebellum as a receptor-free (reference) region was performed to define the most suitable method for regional- and voxel-based quantification of the binding potential (BPND). Finally, standardised uptake value ratio (SUVR-1) was assessed as a potential simplified measurement. RESULTS: For all models, we measured a significant decline in dopamine D1R density (e.g. SRTM = - 38.5 ± 5.0 %, p < 0.0001) in HET mice compared to WT littermates. Shortening the 90-min scan duration resulted in large underestimation of striatal BPND in both WT mice (SRTM 60 min: - 17.7 ± 2.8 %, p = 0.0078) and diseased HET (SRTM 60 min: - 13.1 ± 4.1 %, p = 0.0001). Striatal BPND measurements were very reproducible with an average test-retest variability below 5 % when using both MRTM and SRTM. Parametric BPND maps generated with SRTM were highly reliable, showing nearly perfect agreement to the regional analysis (r2 = 0.99, p < 0.0001). Finally, SRTM provided the most accurate estimate for relative tracer delivery R1 with both regional- and voxel-based analyses. SUVR-1 at different time intervals were not sufficiently reliable when compared to BPND (r2 < 0.66). CONCLUSIONS: Ninety-minute acquisition and the use of SRTM for pharmacokinetic modelling is recommended. [11C]SCH23390 PET imaging demonstrates optimal characteristics for the study of dopamine D1R density in models of psychiatric and neurological disorders as exemplified in the Q175DN mouse model of HD.


Subject(s)
Benzazepines/pharmacokinetics , Brain/diagnostic imaging , Huntington Disease/diagnostic imaging , Molecular Imaging/methods , Positron-Emission Tomography/methods , Receptors, Dopamine D1/antagonists & inhibitors , Animals , Brain/metabolism , Carbon Radioisotopes , Disease Models, Animal , Gene Knock-In Techniques , Huntington Disease/metabolism , Huntington Disease/pathology , Male , Mice , Mice, Transgenic , Receptors, Dopamine D1/metabolism , Reproducibility of Results , Tissue Distribution
16.
Biomed Res Int ; 2021: 4117800, 2021.
Article in English | MEDLINE | ID: mdl-38617025

ABSTRACT

This study is aimed at exploring the effect of pulmonary surfactant (PS) combined with noninvasive positive pressure ventilation on the levels of Keratin-14 (KRT-14) and Endothelin-1 (ET-1) in peripheral blood and the therapeutic effect of neonatal respiratory distress syndrome (NRDS). Altogether 137 cases of neonates with respiratory distress syndrome treated in our hospital from April 2016 to July 2018 were collected. Among them, 64 cases treated with noninvasive positive pressure ventilation were considered as the control group, and 73 cases treated with PS combined with noninvasive positive pressure ventilation were considered as the observation group. The expression of KRT-14 and ET-1 in the two groups was compared. The therapeutic effect, death, complications, and blood gas indexes PaO2, PaCO2, and PaO2/FiO2 in the two groups were compared. Receiver operating characteristic curve (ROC) was applied to analyze the diagnostic value of KRT-14 and ET-1 in the therapeutic effect of NRDS. The effective rate of the observation group was higher than that of the control group. After treatment, PaO2 and PaO2/FiO2 in both groups were notably higher than that before treatment, while PaCO2 was notably lower than that before treatment. And after treatment, the levels of PaO2 and PaO2/FiO2 in the observation group were remarkably higher than that in the control group; PaCO2 was notably lower than that in the control group. After treatment, the levels of KRT-14 and ET-1 in the two groups were remarkably lower than those before treatment, and the levels of KRT-14 and ET-1 in the observation group were considerably lower than those in the control group after treatment. ROC curve showed that the area under the curve (AUC) of KRT-14 was 0.791, and the AUC of ET-1 was 0.816. PS combined with noninvasive positive pressure ventilation can notably improve the therapeutic effect of NRDS. KRT-14 and ET-1 levels may be potential therapeutic diagnostic indicators.

17.
J Med Chem ; 63(15): 8608-8633, 2020 08 13.
Article in English | MEDLINE | ID: mdl-32662649

ABSTRACT

Mutant huntingtin (mHTT) protein carrying the elongated N-terminal polyglutamine (polyQ) tract misfolds and forms protein aggregates characteristic of Huntington's disease (HD) pathology. A high-affinity ligand specific for mHTT aggregates could serve as a positron emission tomography (PET) imaging biomarker for HD therapeutic development and disease progression. To identify such compounds with binding affinity for polyQ aggregates, we embarked on systematic structural activity studies; lead optimization of aggregate-binding affinity, unbound fractions in brain, permeability, and low efflux culminated in the discovery of compound 1, which exhibited target engagement in autoradiography (ARG) studies in brain slices from HD mouse models and postmortem human HD samples. PET imaging studies with 11C-labeled 1 in both HD mice and WT nonhuman primates (NHPs) demonstrated that the right-hand-side labeled ligand [11C]-1R (CHDI-180R) is a suitable PET tracer for imaging of mHTT aggregates. [11C]-1R is now being advanced to human trials as a first-in-class HD PET radiotracer.


Subject(s)
Huntingtin Protein/analysis , Huntington Disease/diagnostic imaging , Positron-Emission Tomography/methods , Protein Aggregation, Pathological/diagnostic imaging , Animals , Disease Models, Animal , Dogs , Female , Humans , Huntingtin Protein/genetics , Huntington Disease/genetics , Ligands , Madin Darby Canine Kidney Cells , Male , Mice , Mice, Inbred C57BL , Mutation , Peptides/genetics , Protein Aggregation, Pathological/genetics , Radiopharmaceuticals/analysis , Rats, Sprague-Dawley
18.
Bioorg Med Chem Lett ; 30(14): 127240, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32527542

ABSTRACT

The (Z)-fluoro-olefin amide bioisosteric replacement is an effective tool for addressing various shortcomings of the parent amide. In an effort to fine tune ADME properties of BACE1 preclinical candidate AM-6494, a series of structurally distinct (Z)-fluoro-olefin containing analogs was developed that culminated in compound 15. Herein, we detail design considerations, synthetic challenges, structure activity relationship (SAR) studies, and in vivo properties of an advanced compound in this novel series of BACE1 inhibitors.


Subject(s)
Alkenes/pharmacology , Amides/pharmacology , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Aspartic Acid Endopeptidases/antagonists & inhibitors , Drug Development , Enzyme Inhibitors/pharmacology , Hydrocarbons, Fluorinated/pharmacology , Alkenes/chemical synthesis , Alkenes/chemistry , Amides/chemical synthesis , Amides/chemistry , Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , HEK293 Cells , Humans , Hydrocarbons, Fluorinated/chemical synthesis , Hydrocarbons, Fluorinated/chemistry , Molecular Conformation , Stereoisomerism , Structure-Activity Relationship
19.
Mol Neurobiol ; 57(4): 2038-2047, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31912442

ABSTRACT

Impairment of group I metabotropic glutamate receptors (mGluRs) results in altered glutamate signalling, which is associated with several neurological disorders including Huntington's Disease (HD), an autosomal neurodegenerative disease. In this study, we assessed in vivo pathological changes in mGluR1 availability in the Q175DN mouse model of HD using longitudinal positron emission tomography (PET) imaging with the radioligand [11C]ITDM. Ninety-minute dynamic PET imaging scans were performed in 22 heterozygous (HET) Q175DN mice and 22 wild-type (WT) littermates longitudinally at 6, 12, and 16 months of age. Analyses of regional volume of distribution with an image-derived input function (VT (IDIF)) and voxel-wise parametric VT (IDIF) maps were performed to assess differences between genotypes. Post-mortem evaluation at 16 months was done to support in vivo findings. [11C]ITDM VT (IDIF) quantification revealed higher mGluR1 availability in the brain of HET mice compared to WT littermates (e.g. cerebellum: + 15.0%, + 17.9%, and + 17.6% at 6, 12, and 16 months, respectively; p < 0.001). In addition, an age-related decline in [11C]ITDM binding independent of genotype was observed between 6 and 12 months. Voxel-wise analysis of parametric maps and post-mortem quantifications confirmed the elevated mGluR1 availability in HET mice compared to WT littermates. In conclusion, in vivo measurement of mGluR1 availability using longitudinal [11C]ITDM PET imaging demonstrated higher [11C]ITDM binding in extra-striatal brain regions during the course of disease in the Q175DN mouse model.


Subject(s)
Huntington Disease/diagnostic imaging , Huntington Disease/metabolism , Positron-Emission Tomography , Receptors, Metabotropic Glutamate/metabolism , Animals , Benzamides , Disease Models, Animal , Mice, Inbred C57BL , Thiazoles
20.
J Cereb Blood Flow Metab ; 40(6): 1351-1362, 2020 06.
Article in English | MEDLINE | ID: mdl-31307287

ABSTRACT

Synaptic pathology is associated with several brain disorders, thus positron emission tomography (PET) imaging of synaptic vesicle glycoprotein 2A (SV2A) using the radioligand [11C]UCB-J may provide a tool to measure synaptic alterations. Given the pivotal role of mouse models in understanding neuropsychiatric and neurodegenerative disorders, this study aims to validate and characterize [11C]UCB-J in mice. We performed a blocking study to verify the specificity of the radiotracer to SV2A, examined kinetic models using an image-derived input function (IDIF) for quantification of the radiotracer, and investigated the in vivo metabolism. Regional TACs during baseline showed rapid uptake of [11C]UCB-J into the brain. Pretreatment with levetiracetam confirmed target engagement in a dose-dependent manner. VT (IDIF) values estimated with one- and two-tissue compartmental models (1TCM and 2TCM) were highly comparable (r=0.999, p < 0.0001), with 1TCM performing better than 2TCM for K1 (IDIF). A scan duration of 60 min was sufficient for reliable VT (IDIF) and K1 (IDIF) estimations. In vivo metabolism of [11C]UCB-J was relatively rapid, with a parent fraction of 22.5 ± 4.2% at 15 min p.i. In conclusion, our findings show that [11C]UCB-J selectively binds to SV2A with optimal kinetics in the mouse representing a promising tool to noninvasively quantify synaptic density in comparative or therapeutic studies in neuropsychiatric and neurodegenerative disorder models.


Subject(s)
Brain/metabolism , Image Processing, Computer-Assisted/methods , Positron-Emission Tomography/methods , Pyridines/pharmacokinetics , Pyrrolidinones/pharmacokinetics , Synaptic Vesicles/metabolism , Animals , Kinetics , Male , Membrane Glycoproteins/analysis , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Models, Theoretical , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/metabolism , Radiopharmaceuticals/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...