20 research outputs found
Structure-Based Design and Synthesis of Benzothiazole Phosphonate Analogues with Inhibitors of Human ABAD-Aβ for Treatment of Alzheimer’s disease
This is the peer reviewed version of the following article: Valasani, K. R., Hu, G., Chaney, M. O. and Yan, S. S. (2013), Structure-Based Design and Synthesis of Benzothiazole Phosphonate Analogues with Inhibitors of Human ABAD-Aβ for Treatment of Alzheimer’s Disease. Chemical Biology & Drug Design, 81: 238–249. doi:10.1111/cbdd.12068, which has been published in final form at http://doi.org/10.1111/cbdd.12068. This article may be used for non-commercial purposes in accordance with Wiley Terms and Conditions for Self-Archiving.Amyloid binding alcohol dehydrogenase (ABAD), a mitochondrial protein, is a cofactor facilitating amyloid-β peptide (Aβ) induced cell stress. Antagonizing Aβ-ABAD interaction protects against aberrant mitochondrial and neuronal function and improves learning memory in the Alzheimer’s disease (AD) mouse model. Therefore, it offers a potential target for Alzheimer’s drug design, by identifying potential inhibitors of Aβ-ABAD interaction. 2D QSAR methods were applied to novel compounds with known IC50 values, which formed a training set. A correlation analysis was carried out comparing the statistics of the measured IC50 with predicted values. These selectivity-determining descriptors were interpreted graphically in terms of principle component analyses, which are highly informative for the lead optimization process with respect to activity enhancement. A 3D pharmacophore model also was created. The 2D QSAR and 3D pharmacophore models will assist in hi-throughput screening. In addition, ADME descriptors were also determined to study their pharmacokinetic properties. Finally, ABAD molecular docking study of these novel molecules was undertaken to determine whether these compounds exhibit significant binding affinity with the binding site. We have synthesized only the compounds that have shown the best drug like properties as candidates for further studies
Mitochondrial permeability transition pore is a potential drug target for neurodegeneration
Mitochondrial permeability transition pore (mPTP) plays a central role in alterations of mitochondrial structure and function leading to neuronal injury relevant to aging and neurodegenerative diseases including Alzheimer’s disease (AD). mPTP putatively consists of the voltage-dependent anion channel (VDAC) and the adenine nucleotide translocator (ANT). Cyclophilin D (CypD) and reactive oxygen species (ROS) increase intra-cellular calcium and enhance the formation of mPTP that leads to neuronal cell death in AD. CypD-dependent mPTP can play a crucial role in ischemia/reperfusion injury. The interaction of amyloid beta peptide (Aβ) with CypD potentiates mitochondrial and neuronal perturbation. This interaction triggers the formation of mPTP, resulting in decreased mitochondrial membrane potential, impaired mitochondrial respiration function, increased oxidative stress, release of cytochrome c, and impaired axonal mitochondrial transport. Thus, the CypD-dependent mPTP is directly linked to the cellular and synaptic perturbations observed in the pathogenesis of AD. Designing small molecules to block this interaction would lessen the effects of Aβ neurotoxicity. This review summarizes the recent progress on mPTP and its potential therapeutic target for neurodegenerative diseases including AD
From a Cell’s Viewpoint: Targeting Mitochondria in Alzheimer’s disease
Mitochondria are well-known cellular organelles widely studied in relation to a variety of disease states, including Alzheimer’s disease. With roles in several metabolic processes, numerous signal transduction pathways, and overall cell maintenance and survival, mitochondria are essential to understanding the inner workings of cells. As mitochondria are able to be utilized by diverse illnesses to increase the likelihood of disease progression, targeting specific processes in these organelles could provide beneficial therapeutic options
Mitochondrial permeability transition pore is a potential drug target for neurodegeneration
Mitochondrial permeability transition pore (mPTP) plays a central role in alterations of mitochondrial structure and function leading to neuronal injury relevant to aging and neurodegenerative diseases including Alzheimer’s disease (AD). mPTP putatively consists of the voltage-dependent anion channel (VDAC) and the adenine nucleotide translocator (ANT). Cyclophilin D (CypD) and reactive oxygen species (ROS) increase intra-cellular calcium and enhance the formation of mPTP that leads to neuronal cell death in AD. CypD-dependent mPTP can play a crucial role in ischemia/reperfusion injury. The interaction of amyloid beta peptide (Aβ) with CypD potentiates mitochondrial and neuronal perturbation. This interaction triggers the formation of mPTP, resulting in decreased mitochondrial membrane potential, impaired mitochondrial respiration function, increased oxidative stress, release of cytochrome c, and impaired axonal mitochondrial transport. Thus, the CypD-dependent mPTP is directly linked to the cellular and synaptic perturbations observed in the pathogenesis of AD. Designing small molecules to block this interaction would lessen the effects of Aβ neurotoxicity. This review summarizes the recent progress on mPTP and its potential therapeutic target for neurodegenerative diseases including AD
Identification of human presequence protease (hPreP) agonists for the treatment of Alzheimer’s disease
Amyloid-β (Aβ), a neurotoxic peptide, is linked to the onset of Alzheimer’s disease (AD). Increased Aβ content within neuronal cell mitochondria is a pathological feature in both human and mouse models with AD. This accumulation of Aβ within the mitochondrial landscape perpetuates increased free radical production and activation of the apoptotic pathway. Human Presequence Protease (hPreP) is responsible for the degradation of mitochondrial amyloid-β peptide in human neuronal cells, and is thus an attractive target to increase the proteolysis of Aβ. Therefore, it offers a potential target for Alzheimer’s drug design, by identifying potential activators of hPreP. We applied structure-based drug design, combined with experimental methodologies to investigate the ability of various compounds to enhance hPreP proteolytic activity. Compounds 3c & 4c enhanced hPreP-mediated proteolysis of Aβ (1–42), pF1β (2–54) and fluorogenic-substrate V. These results suggest that activation of hPreP by small benzimidazole derivatives provide a promising avenue for AD treatment
Synthesis and bioactivity of phosphorylated derivatives of stavudine
Novel phosphorylated derivatives of stavudine were synthesized by the reaction of bis(2-chloroethyl)phosphoramidic dichloride/4-nitrophenyl phosphorodichloridate with various cyclic amines and amino acid esters in the presence of triethylamine in dry tetrahydrofuran through the corresponding monochloride intermediates 2a-l. Further reaction of the intermediates 2a-l with stavudine in tetrahydrofuran and pyridine in the presence of triethylamine formed the title compounds 4a-l. Their structures were characterized by IR, Â Â 1H-, 13C-, 31P-NMR and mass spectral data analyses. They exhibited good antibacterial and antioxidant activities. Their bioactivity was greatly influenced by the different groups present at the phosphorus
Determination of Small Molecule ABAD Inhibitors Crossing Blood Brain Barrier and Pharmacokinetics
A major obstacle to the development of effective treatment of Alzheimer’s disease (AD) is successfully delivery of drugs to the brain. We have previously identified a series of benzothiazole phosphonate compounds that block the interaction of amyloid beta peptide (Aβ) with amyloid-beta binding alcohol dehydrogenase (ABAD). A selective and sensitive method for the presence of three new benzothiazole ABAD inhibitors in mouse plasma, brain and artificial cerebrospinal fluid has been developed and validated based on high performance liquid chromatography tandem mass spectrometry. Mass spectra were generated using Micromass Quattro Ultima “triple” quadrupole mass spectrometer equipped with Electrospray ionization interface. Good linearity was obtained over a concentration range of 0.05–2.5 µg/ml. The lowest limit of quantification and detection was 0.05µg/ml. All inter-day accuracies and precisions were within ±15% of the nominal value and ±20%, respectively, at the lower limit of quantitation. The tested compounds were stable at various conditions with recoveries >90.0 % (RSD<10%). The method used for pharmacokinetic studies of compounds in mouse cerebrospinal fluid, plasma, and brain is accurate, precise, and specific with no matrix effect. Pharmacokinetic data showed these compounds penetrate the blood–brain barrier (BBB) yielding 4–50 ng/ml peak brain concentrations and 2 µg/ml peak plasma concentrations from a 10mg/kg dose. These results indicate that our newly synthesized small molecule ABAD inhibitor have good drug properties with the ability to cross the blood brain barrier, which holds a great potential for AD therapy
Identification of Human ABAD Inhibitors for Rescuing Aβ-Mediated Mitochondrial Dysfunction
Amyloid beta (Aβ) binding alcohol dehydrogenase (ABAD) is a cellular cofactor for promoting (Aβ)-mediated mitochondrial and neuronal dysfunction, and cognitive decline in transgenic Alzheimer's disease (AD) mouse models. Targeting mitochondrial ABAD may represent a novel therapeutic strategy against AD. Here, we report the biological activity of small molecule ABAD inhibitors. Using in vitro surface plasmon resonance (SPR) studies, we synthesized compounds with strong binding affinities for ABAD. Further, these ABAD inhibitors (ABAD-4a and 4b) reduced ABAD enzyme activity and administration of phosphonate derivatives of ABAD inhibitors antagonized calcium-mediated mitochondrial swelling. Importantly, these compounds also abolished Aβ-induced mitochondrial dysfunction as shown by increased cytochrome c oxidase and adenosine-5′-triphosphate levels, suggesting protective mitochondrial function effects of these synthesized compounds. Thus, these compounds are potential candidates for further pharmacologic development to target ABAD to improve mitochondrial function
First stereoselective total synthesis of (3<i style="">R</i>, 3a<i style="">S</i>, 6a<i style="">R</i>)-hexahydrofuro[2,3-<i>b</i>]furan-3-yl (2<i style="">R</i>,3<i style="">S</i>)-4-(4-amino-N-isobutyl phenylsulfonamido)-3-hydroxy-1-phenylbutan-2-yl-carbamate (diastereomer of Darunavir)
849-854The stereoselective
novel synthesis of (3R,3aS,6aR)-hexahydrofuro
[2,3-b]furan-3-yl (2R,3S)-4-(4-amino-N-isobutyl
phenyl sulfonamido)-3-hydroxy-1-phenylbutan-2-yl-carbamate, has been
accomplished in situ from the intermediate 4-amino-N-((2S,3R)-3-amino-2-hydroxy-4-phenylbutyl)-N-isobutylbenzen
sulfonamide 7. This reaction gives 88% yield, with an optical purity of
99.5% and diastereomeric purity of 99% for chiral alcohol 8
Acetylcholinesterase Inhibitors: Structure Based Design, Synthesis, Pharmacophore Modeling, and Virtual Screening
Acetylcholinesterase (AChE) is a
main drug target, and its inhibitors
have demonstrated functionality in the symptomatic treatment of Alzheimer’s
disease (AD). In this study, a series of novel AChE inhibitors were
designed and their inhibitory activity was evaluated with 2D quantitative
structure–activity relationship (QSAR) studies using a training
set of 20 known compounds for which IC<sub>50</sub> values had previously
been determined. The QSAR model was calculated based on seven unique
descriptors. Model validation was determined by predicting IC<sub>50</sub> values for a test set of 20 independent compounds with measured
IC<sub>50</sub> values. A correlation analysis was carried out comparing
the statistics of the measured IC<sub>50</sub> values with predicted
ones. These selectivity-determining descriptors were interpreted graphically
in terms of principal component analyses (PCA). A 3D pharmacophore
model was also created based on the activity of the training set.
In addition, absorption, distribution, metabolism, and excretion (ADME)
descriptors were also determined to evaluate their pharmacokinetic
properties. Finally, molecular docking of these novel molecules into
the AChE binding domain indicated that three molecules (<b>6c</b>, <b>7c</b>, and <b>7h)</b> should have significantly
higher affinities and solvation energies than the known standard drug
donepezil. The docking studies of 2<i>H</i>-thiazoloÂ[3,2-<i>a</i>]Âpyrimidines (<b>6a</b>–<b>6j</b>) and
5<i>H</i>-thiazoloÂ[3,2-<i>a</i>] pyrimidines (<b>7a</b>–<b>7j</b>) with human AChE have demonstrated
that these ligands bind to the dual sites of the enzyme. Simple and
ecofriendly syntheses and diastereomeric crystallizations of 2<i>H</i>-thiazolo [3,2-<i>a</i>]Âpyrimidines and 5<i>H</i>-thiazoloÂ[3,2-<i>a</i>] pyrimidines are described.
The solid-state structures for the HBr salts of compounds <b>6a</b>,<b> 6e</b>, <b>7a</b>, and <b>7i</b> have been
determined using single-crystal X-ray diffraction techniques, and
X-ray powder patterns were measured for the bulk solid remaining after
solvent was removed from solutions containing <b>6a</b> and <b>7a</b>. These studies provide valuable insight for designing more
potent and selective inhibitors for the treatment of AD