321 results on '"Silverman RB"'
Search Results
2. Synthesis of (2 R ,4 S )-4-Amino-5-hydroxybicyclo[3.1.1]heptane-2-carboxylic Acid via an Asymmetric Intramolecular Mannich Reaction.
- Author
-
Dukes AO, Weerawarna PM, Devitt AN, and Silverman RB
- Subjects
- Stereoisomerism, Molecular Structure, Bridged Bicyclo Compounds chemistry, Bridged Bicyclo Compounds chemical synthesis, Humans, Carboxylic Acids chemistry
- Abstract
Inhibition of human ornithine aminotransferase interferes with glutamine and proline metabolism in hepatocellular carcinoma, depriving tumors of essential nutrients. A proposed mechanism-based inhibitor containing a bicyclo[3.1.1]heptanol warhead is reported herein. The proposed inactivation mechanism involves a novel α-iminol rearrangement. The synthesis of the proposed inhibitor features an asymmetric intramolecular Mannich reaction, utilizing a chiral sulfinamide. This study presents a novel approach toward the synthesis of functionalized bicyclo[3.1.1]heptanes and highlights an underutilized method to access enantiopure exocyclic amines.
- Published
- 2024
- Full Text
- View/download PDF
3. Design, Synthesis, and Mechanistic Studies of ( R )-3-Amino-5,5-difluorocyclohex-1-ene-1-carboxylic Acid as an Inactivator of Human Ornithine Aminotransferase.
- Author
-
Devitt AN, Vargas AL, Zhu W, Des Soye BJ, Butun FA, Alt T, Kaley N, Ferreira GM, Moran GR, Kelleher NL, Liu D, and Silverman RB
- Subjects
- Humans, Carboxylic Acids chemistry, Carboxylic Acids chemical synthesis, Carboxylic Acids pharmacology, Cyclohexenes chemistry, Cyclohexenes chemical synthesis, Cyclohexenes pharmacology, Cyclohexenes metabolism, Carcinoma, Hepatocellular drug therapy, Carcinoma, Hepatocellular metabolism, Crystallography, X-Ray, Models, Molecular, Ornithine-Oxo-Acid Transaminase metabolism, Ornithine-Oxo-Acid Transaminase chemistry, Ornithine-Oxo-Acid Transaminase antagonists & inhibitors, Drug Design, Enzyme Inhibitors pharmacology, Enzyme Inhibitors chemistry, Enzyme Inhibitors chemical synthesis
- Abstract
Human ornithine aminotransferase ( h OAT), a pyridoxal 5'-phosphate (PLP)-dependent enzyme, has been shown to play an essential role in the metabolic reprogramming and progression of hepatocellular carcinoma (HCC). HCC accounts for approximately 75% of primary liver cancers and is within the top three causes of cancer death worldwide. As a result of treatment limitations, the overall 5-year survival rate for all patients with HCC is under 20%. The prevalence of HCC necessitates continued development of novel and effective treatment methods. In recent years, the therapeutic potential of selective inactivation of h OAT has been demonstrated for the treatment of HCC. Inspired by previous increased selectivity for h OAT by the expansion of the cyclopentene ring scaffold to a cyclohexene, we designed, synthesized, and evaluated a series of novel fluorinated cyclohexene analogues and identified (R)-3-amino-5,5-difluorocyclohex-1-ene-1-carboxylic acid as a time-dependent inhibitor of h OAT. Structural and mechanistic studies have elucidated the mechanism of inactivation of h OAT by 5 , resulting in a PLP-inactivator adduct tightly bound to the active site of the enzyme. Intact protein mass spectrometry,
19 F NMR spectroscopy, transient state kinetic studies, and X-ray crystallography were used to determine the structure of the final adduct and elucidate the mechanisms of inactivation. Interestingly, despite the highly electrophilic intermediate species conferred by fluorine and structural evidence of solvent accessibility in the h OAT active site, Lys292 and water did not participate in nucleophilic addition during the inactivation mechanism of h OAT by 5 . Instead, rapid aromatization to yield the final adduct was favored.- Published
- 2024
- Full Text
- View/download PDF
4. Crystallographic and Computational Insights into Isoform-Selective Dynamics in Nitric Oxide Synthase.
- Author
-
Li H, Hardy CD, Reidl CT, Jing Q, Xue F, Cinelli M, Silverman RB, and Poulos TL
- Subjects
- Nitric Oxide Synthase Type I, Protein Isoforms chemistry, Crystallography, X-Ray, Enzyme Inhibitors pharmacology, Heme chemistry, Tyrosine, Nitric Oxide, Nitric Oxide Synthase metabolism, Nitric Oxide Synthase Type III genetics, Nitric Oxide Synthase Type III chemistry
- Abstract
In our efforts to develop inhibitors selective for neuronal nitric oxide synthase (nNOS) over endothelial nitric oxide synthase (eNOS), we found that nNOS can undergo conformational changes in response to inhibitor binding that does not readily occur in eNOS. One change involves movement of a conserved tyrosine, which hydrogen bonds to one of the heme propionates, but in the presence of an inhibitor, changes conformation, enabling part of the inhibitor to hydrogen bond with the heme propionate. This movement does not occur as readily in eNOS and may account for the reason why these inhibitors bind more tightly to nNOS. A second structural change occurs upon the binding of a second inhibitor molecule to nNOS, displacing the pterin cofactor. Binding of this second site inhibitor requires structural changes at the dimer interface, which also occurs more readily in nNOS than in eNOS. Here, we used a combination of crystallography, mutagenesis, and computational methods to better understand the structural basis for these differences in NOS inhibitor binding. Computational results show that a conserved tyrosine near the primary inhibitor binding site is anchored more tightly in eNOS than in nNOS, allowing for less flexibility of this residue. We also find that the inefficiency of eNOS to bind a second inhibitor molecule is likely due to the tighter dimer interface in eNOS compared with nNOS. This study provides a better understanding of how subtle structural differences in NOS isoforms can result in substantial dynamic differences that can be exploited in the development of isoform-selective inhibitors.
- Published
- 2024
- Full Text
- View/download PDF
5. Target Identification of a Class of Pyrazolone Protein Aggregation Inhibitor Therapeutics for Amyotrophic Lateral Sclerosis.
- Author
-
Weerawarna PM, Schiefer IT, Soares P, Fox S, Morimoto RI, Melani RD, Kelleher NL, Luan CH, and Silverman RB
- Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease with no cure, and current treatment options are very limited. Previously, we performed a high-throughput screen to identify small molecules that inhibit protein aggregation caused by a mutation in the gene that encodes superoxide dismutase 1 (SOD1), which is responsible for about 25% of familial ALS. This resulted in three hit series of compounds that were optimized over several years to give three compounds that were highly active in a mutant SOD1 ALS model. Here we identify the target of two of the active compounds ( 6 and 7 ) with the use of photoaffinity labeling, chemical biology reporters, affinity purification, proteomic analysis, and fluorescent/cellular thermal shift assays. Evidence is provided to demonstrate that these two pyrazolone compounds directly interact with 14-3-3-E and 14-3-3-Q isoforms, which have chaperone activity and are known to interact with mutant SOD1
G93A aggregates and become insoluble in the subcellular JUNQ compartment, leading to apoptosis. Because protein aggregation is the hallmark of all neurodegenerative diseases, knowledge of the target compounds that inhibit protein aggregation allows for the design of more effective molecules for the treatment of ALS and possibly other neurodegenerative diseases., Competing Interests: The authors declare no competing financial interest., (© 2023 The Authors. Published by American Chemical Society.)- Published
- 2023
- Full Text
- View/download PDF
6. From Tg O/GABA-AT, GABA, and T-263 Mutant to Conception of Toxoplasma .
- Author
-
Lykins J, Moschitto MJ, Zhou Y, Filippova EV, Le HV, Tomita T, Fox BA, Bzik DJ, Su C, Rajagopala SV, Flores K, Spano F, Woods S, Roberts CW, Hua C, El Bissati K, Wheeler KM, Dovgin S, Muench SP, McPhillie M, Fishwick CWG, Anderson WF, Lee PJ, Hickman M, Weiss LM, Dubey JP, Lorenzi HA, Silverman RB, and McLeod RL
- Abstract
Toxoplasma gondii causes morbidity, mortality, and disseminates widely via cat sexual stages. Here, we find T. gondii ornithine aminotransferase (OAT) is conserved across phyla. We solve Tg O/GABA-AT structures with bound inactivators at 1.55 Å and identify an inactivator selective for Tg O/GABA-AT over human OAT and GABA-AT. However, abrogating Tg O/GABA-AT genetically does not diminish replication, virulence, cyst-formation, or eliminate cat's oocyst shedding. Increased sporozoite/merozoite Tg O/GABA-AT expression led to our study of a mutagenized clone with oocyst formation blocked, arresting after forming male and female gametes, with "Rosetta stone"-like mutations in genes expressed in merozoites. Mutations are similar to those in organisms from plants to mammals, causing defects in conception and zygote formation, affecting merozoite capacitation, pH/ionicity/sodium-GABA concentrations, drawing attention to cyclic AMP/PKA, and genes enhancing energy or substrate formation in Tg O/GABA-AT-related-pathways. These candidates potentially influence merozoite's capacity to make gametes that fuse to become zygotes, thereby contaminating environments and causing disease., Competing Interests: H.V.L. is currently a program officer at the National Institute on Drug Abuse at the National Institutes of Health. H.A.L. is currently a scientist working at National Institute of Diabetes, Digestive and Kidney Disease (NIDDK) at the National Institutes of Health. The findings and conclusions of this article are those of the authors and do not necessarily reflect the views of the National Institute on Drug Abuse, National Institute of Diabetes, Digestive and Kidney Diseases (NIDDK), the National Institutes of Health, nor the US Department of Health and Human Services. R.B.S. and R.L.M., with H.V.L., submitted Patent Number: US 10,632,088 B2 INACTIVATORS OF TOXOPLASMA GONDII ORNITHINE AMINOTRANSFERASE FOR TREATING TOXOPLASMOSIS AND MALARIA through their Technology Transfer offices. The authors declare no competing financial interest., (© 2023 The Authors.)
- Published
- 2023
- Full Text
- View/download PDF
7. Small molecules targeting different cellular pathologies for the treatment of amyotrophic lateral sclerosis.
- Author
-
Elmansy MF, Reidl CT, Rahaman M, Özdinler PH, and Silverman RB
- Subjects
- Animals, Humans, Disease Models, Animal, Motor Neurons metabolism, Motor Neurons pathology, Amyotrophic Lateral Sclerosis drug therapy, Amyotrophic Lateral Sclerosis genetics, Neurodegenerative Diseases metabolism
- Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease in which the motor neuron circuitry displays progressive degeneration, affecting mostly the motor neurons in the brain and in the spinal cord. There are no effective cures, albeit three drugs, riluzole, edaravone, and AMX0035 (a combination of sodium phenylbutyrate and taurursodiol), have been approved by the Food and Drug Administration, with limited improvement in patients. There is an urgent need to build better and more effective treatment strategies for ALS. Since the disease is very heterogenous, numerous approaches have been explored, such as targeting genetic mutations, decreasing oxidative stress and excitotoxicity, enhancing mitochondrial function and protein degradation mechanisms, and inhibiting neuroinflammation. In addition, various chemical libraries or previously identified drugs have been screened for potential repurposing in the treatment of ALS. Here, we review previous drug discovery efforts targeting a variety of cellular pathologies that occur from genetic mutations that cause ALS, such as mutations in SOD1, C9orf72, FUS, and TARDP-43 genes. These mutations result in protein aggregation, which causes neuronal degeneration. Compounds used to target cellular pathologies that stem from these mutations are discussed and comparisons among different preclinical models are presented. Because the drug discovery landscape for ALS and other motor neuron diseases is changing rapidly, we also offer recommendations for a novel, more effective, direction in ALS drug discovery that could accelerate translation of effective compounds from animals to patients., (© 2023 The Authors. Medicinal Research Reviews published by Wiley Periodicals LLC.)
- Published
- 2023
- Full Text
- View/download PDF
8. Potent, Selective, and Membrane Permeable 2-Amino-4-Substituted Pyridine-Based Neuronal Nitric Oxide Synthase Inhibitors.
- Author
-
Vasu D, Do HT, Li H, Hardy CD, Awasthi A, Poulos TL, and Silverman RB
- Subjects
- Rats, Mice, Humans, Animals, Nitric Oxide Synthase Type I, Structure-Activity Relationship, Nitric Oxide, Nitric Oxide Synthase chemistry, Nitric Oxide Synthase metabolism, Enzyme Inhibitors pharmacology, Enzyme Inhibitors chemistry
- Abstract
A series of potent, selective, and highly permeable human neuronal nitric oxide synthase inhibitors (hnNOS), based on a difluorobenzene ring linked to a 2-aminopyridine scaffold with different functionalities at the 4-position, is reported. In our efforts to develop novel nNOS inhibitors for the treatment of neurodegenerative diseases, we discovered 17 , which showed excellent potency toward both rat ( K
i 15 nM) and human nNOS ( Ki 19 nM), with 1075-fold selectivity over human eNOS and 115-fold selectivity over human iNOS. 17 also showed excellent permeability ( Pe = 13.7 × 10-6 cm s-1 ), a low efflux ratio (ER 0.48), along with good metabolic stability in mouse and human liver microsomes, with half-lives of 29 and >60 min, respectively. X-ray cocrystal structures of inhibitors bound with three NOS enzymes, namely, rat nNOS, human nNOS, and human eNOS, revealed detailed structure-activity relationships for the observed potency, selectivity, and permeability properties of the inhibitors.- Published
- 2023
- Full Text
- View/download PDF
9. Improved synthesis and anticancer activity of a potent neuronal nitric oxide synthase inhibitor.
- Author
-
Vasu D, Reidl CT, Wang E, Yang S, and Silverman RB
- Subjects
- Mice, Humans, Animals, Nitric Oxide Synthase Type I, Enzyme Inhibitors pharmacology, Cells, Cultured, Nitric Oxide, Nitric Oxide Synthase, Melanoma
- Abstract
An improved synthesis of 4-methyl-7-(3-((methylamino)methyl)phenethyl)quinolin-2-amine (1) is reported. A scalable, rapid, and efficient methodology was developed to access this compound with an overall yield of 35%, which is 5.9-fold higher than the previous report. The key differences in the improved synthesis are a high yielding quinoline synthesis by a Knorr reaction, a copper-mediated Sonogashira coupling to the internal alkyne in excellent yield, and a crucial deprotection of the N-acetyl and N-Boc groups achieved under acidic conditions in a single step rather than a poor yielding quinoline N-oxide strategy, basic deprotection conditions, and low yielding copper-free conditions that were reported in the previous report. Compound 1, which previously was shown to inhibit IFN-γ-induced tumor growth in a human melanoma xenograft mouse model, was found to inhibit the growth of metastatic melanoma, glioblastoma, and hepatocellular carcinoma in vitro., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2023 Elsevier Ltd. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
10. Correction to "Rational Design, Synthesis, and Mechanism of (3 S ,4 R )-3-Amino-4-(difluoromethyl)cyclopent-1-ene-1-carboxylic Acid: Employing a Second-Deprotonation Strategy for Selectivity of Human Ornithine Aminotransferase over GABA Aminotransferase".
- Author
-
Zhu W, Butrin A, Melani RD, Doubleday PF, Ferreira GM, Tavares MT, Habeeb Mohammad TS, Beaupre BA, Kelleher NL, Moran GR, Liu D, and Silverman RB
- Published
- 2023
- Full Text
- View/download PDF
11. Structural and Mechanistic Basis for the Inactivation of Human Ornithine Aminotransferase by (3 S ,4 S )-3-Amino-4-fluorocyclopentenecarboxylic Acid.
- Author
-
Shen S, Butrin A, Beaupre BA, Ferreira GM, Doubleday PF, Grass DH, Zhu W, Kelleher NL, Moran GR, Liu D, and Silverman RB
- Subjects
- Humans, Amino Acids pharmacology, Enzyme Inhibitors pharmacology, Ornithine-Oxo-Acid Transaminase chemistry, Ornithine-Oxo-Acid Transaminase metabolism, gamma-Aminobutyric Acid, Carboxylic Acids pharmacology, Carboxylic Acids chemistry, Ornithine, Carcinoma, Hepatocellular, Liver Neoplasms
- Abstract
Ornithine aminotransferase (OAT) is overexpressed in hepatocellular carcinoma (HCC), and we previously showed that inactivation of OAT inhibits the growth of HCC. Recently, we found that (3 S ,4 S )-3-amino-4-fluorocyclopentenecarboxylic acid ( 5 ) was a potent inactivator of γ-aminobutyric acid aminotransferase (GABA-AT), proceeding by an enamine mechanism. Here we describe our investigations into the activity and mechanism of 5 as an inactivator of human OAT. We have found that 5 exhibits 10-fold less inactivation efficiency ( k
inact / KI ) against h OAT than GABA-AT. A comprehensive mechanistic study was carried out to understand its inactivation mechanism with h OAT. p Ka and electrostatic potential calculations were performed to further support the notion that the α,β-unsaturated alkene of 5 is critical for enhancing acidity and nucleophilicity of the corresponding intermediates and ultimately responsible for the improved inactivation efficiency of 5 over the corresponding saturated analogue ( 4 ). Intact protein mass spectrometry and the crystal structure complex with h OAT provide evidence to conclude that 5 mainly inactivates h OAT through noncovalent interactions, and that, unlike with GABA-AT, covalent binding with h OAT is a minor component of the total inhibition which is unique relative to other monofluoro-substituted derivatives. Furthermore, based on the results of transient-state measurements and free energy calculations, it is suggested that the α,β-unsaturated carboxylate group of PLP-bound 5 may be directly involved in the inactivation cascade by forming an enolate intermediate. Overall, compound 5 exhibits unusual structural conversions which are catalyzed by specific residues within h OAT, ultimately leading to an enamine mechanism-based inactivation of h OAT through noncovalent interactions and covalent modification.- Published
- 2023
- Full Text
- View/download PDF
12. 2-Aminopyridines with a shortened amino sidechain as potent, selective, and highly permeable human neuronal nitric oxide synthase inhibitors.
- Author
-
Vasu D, Li H, Hardy CD, Poulos TL, and Silverman RB
- Subjects
- Animals, Enzyme Inhibitors chemistry, Enzyme Inhibitors pharmacology, Humans, Nitric Oxide Synthase, Nitric Oxide Synthase Type I chemistry, Nitric Oxide Synthase Type I metabolism, Protein Isoforms, Rats, Aminopyridines chemistry, Aminopyridines pharmacology, Nitric Oxide
- Abstract
A series of potent, selective, and highly permeable human neuronal nitric oxide synthase inhibitors (hnNOS) based on the 2-aminopyridine scaffold with a shortened amino sidechain is reported. A rapid and simple protocol was developed to access these inhibitors in excellent yields. Neuronal nitric oxide synthase (nNOS) is a novel therapeutic target for the treatment of various neurological disorders. The major challenges in designing nNOS inhibitors in humans focus on potency, selectivity over other isoforms of nitric oxide synthases (NOSs), and blood-brain barrier permeability. In this context, we discovered a promising inhibitor, 6-(3-(4,4-difluoropiperidin-1-yl)propyl)-4-methylpyridin-2-amine dihydrochloride, that exhibits excellent potency for rat (K
i = 46 nM) and human nNOS (Ki = 48 nM), respectively, with 388-fold human eNOS and 135-fold human iNOS selectivity. It also displayed excellent permeability (Pe = 17.3 × 10-6 cm s-1 ) through a parallel artificial membrane permeability assay, a model for blood-brain permeability. We found that increasing lipophilicity by incorporation of fluorine atoms on the backbone of the inhibitors significantly increased potential blood-brain barrier permeability. In addition to measuring potency, isoform selectivity, and permeability of NOS inhibitors, we also explored structure-activity relationships via structures of key inhibitors complexed to various isoforms of nitric oxide synthases., (Copyright © 2022 Elsevier Ltd. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
13. A Small Peptide Increases Drug Delivery in Human Melanoma Cells.
- Author
-
Tong S, Darwish S, Ariani HHN, Lozada KA, Salehi D, Cinelli MA, Silverman RB, Kaur K, and Yang S
- Abstract
Melanoma is the most fatal type of skin cancer and is notoriously resistant to chemotherapies. The response of melanoma to current treatments is difficult to predict. To combat these challenges, in this study, we utilize a small peptide to increase drug delivery to melanoma cells. A peptide library array was designed and screened using a peptide array-whole cell binding assay, which identified KK-11 as a novel human melanoma-targeting peptide. The peptide and its D-amino acid substituted analogue (VPWxEPAYQrFL or D-aa KK-11) were synthesized via a solid-phase strategy. Further studies using FITC-labeled KK-11 demonstrated dose-dependent uptake in human melanoma cells. D-aa KK-11 significantly increased the stability of the peptide, with 45.3% remaining detectable after 24 h with human serum incubation. Co-treatment of KK-11 with doxorubicin was found to significantly enhance the cytotoxicity of doxorubicin compared to doxorubicin alone, or sequential KK-11 and doxorubicin treatment. In vivo and ex vivo imaging revealed that D-aa KK-11 distributed to xenografted A375 melanoma tumors as early as 5 min and persisted up to 24 h post tail vein injection. When co-administered, D-aa KK-11 significantly enhanced the anti-tumor activity of a novel nNOS inhibitor (MAC-3-190) in an A375 human melanoma xenograft mouse model compared to MAC-3-190 treatment alone. No apparent systemic toxicities were observed. Taken together, these results suggest that KK-11 may be a promising human melanoma-targeted delivery vector for anti-melanoma cargo.
- Published
- 2022
- Full Text
- View/download PDF
14. Palladium-Catalyzed α-Arylation of Cyclic β-Dicarbonyl Compounds for the Synthesis of Ca V 1.3 Inhibitors.
- Author
-
Yun J, Jeong D, Xie Z, Lee S, Kim J, Surmeier DJ, Silverman RB, and Kang S
- Abstract
Cyclic α-aryl β-dicarbonyl derivatives are important scaffolds in medicinal chemistry. Palladium-catalyzed coupling reactions of haloarenes were conducted with diverse five- to seven-membered cyclic β-dicarbonyl derivatives including barbiturate, pyrazolidine-3,5-dione, and 1,4-diazepane-5,7-dione. The coupling reactions of various para- or meta-substituted aryl halides occurred efficiently when Pd( t -Bu
3 P)2 , Xphos, and Cs2 CO3 were used under 1,4-dioxane reflux conditions. Although the couplings of ortho-substituted aryl halides with pyrazolidine-3,5-dione and 1,4-diazepane-5,7-dione were moderate, the coupling with barbiturate was limited. Using the optimized reaction conditions, we synthesized several 5-aryl barbiturates as new scaffolds of CaV 1.3 Ca2+ channel inhibitors. Among the synthesized molecules, 14e was the most potent CaV 1.3 inhibitor with an IC50 of 1.42 μM., Competing Interests: The authors declare no competing financial interest., (© 2022 The Authors. Published by American Chemical Society.)- Published
- 2022
- Full Text
- View/download PDF
15. NU-9 improves health of hSOD1 G93A mouse upper motor neurons in vitro, especially in combination with riluzole or edaravone.
- Author
-
Genç B, Gautam M, Helmold BR, Koçak N, Günay A, Goshu GM, Silverman RB, and Hande Ozdinler P
- Subjects
- Animals, Edaravone pharmacology, Humans, Mice, Motor Neurons, Superoxide Dismutase, Amyotrophic Lateral Sclerosis drug therapy, Riluzole pharmacology, Riluzole therapeutic use
- Abstract
Even though amyotrophic lateral sclerosis (ALS) is a disease of the upper and lower motor neurons, to date none of the compounds in clinical trials have been tested for improving the health of diseased upper motor neurons (UMNs). There is an urgent need to develop preclinical assays that include UMN health as a readout. Since ALS is a complex disease, combinatorial treatment strategies will be required to address the mechanisms perturbed in patients. Here, we describe a novel in vitro platform that takes advantage of an UMN reporter line in which UMNs are genetically labeled with fluorescence and have misfolded SOD1 toxicity. We report that NU-9, an analog of the cyclohexane-1,3-dione family of compounds, improves the health of UMNs with misfolded SOD1 toxicity more effectively than riluzole or edaravone, -the only two FDA-approved ALS drugs to date-. Interestingly, when NU-9 is applied in combination with riluzole or edaravone, there is an additive effect on UMN health, as they extend longer axons and display enhanced branching and arborization, two important characteristics of healthy UMNs in vitro., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
16. Rational Design, Synthesis, and Mechanism of (3 S ,4 R )-3-Amino-4-(difluoromethyl)cyclopent-1-ene-1-carboxylic Acid: Employing a Second-Deprotonation Strategy for Selectivity of Human Ornithine Aminotransferase over GABA Aminotransferase.
- Author
-
Zhu W, Butrin A, Melani RD, Doubleday PF, Ferreira GM, Tavares MT, Habeeb Mohammad TS, Beaupre BA, Kelleher NL, Moran GR, Liu D, and Silverman RB
- Subjects
- Carboxylic Acids, Enzyme Inhibitors chemistry, Humans, Kinetics, Molecular Docking Simulation, Ornithine-Oxo-Acid Transaminase, Phenylacetates, Pyridoxal Phosphate chemistry, gamma-Aminobutyric Acid, 4-Aminobutyrate Transaminase, Liver Neoplasms
- Abstract
Human ornithine aminotransferase (hOAT) is a pyridoxal 5'-phosphate (PLP)-dependent enzyme that contains a similar active site to that of γ-aminobutyric acid aminotransferase (GABA-AT). Recently, pharmacological inhibition of hOAT was recognized as a potential therapeutic approach for hepatocellular carcinoma. In this work, we first studied the inactivation mechanisms of hOAT by two well-known GABA-AT inactivators ( CPP-115 and OV329 ). Inspired by the inactivation mechanistic difference between these two aminotransferases, a series of analogues were designed and synthesized, leading to the discovery of analogue 10b as a highly selective and potent hOAT inhibitor. Intact protein mass spectrometry, protein crystallography, and dialysis experiments indicated that 10b was converted to an irreversible tight-binding adduct ( 34 ) in the active site of hOAT, as was the unsaturated analogue ( 11 ). The comparison of kinetic studies between 10b and 11 suggested that the active intermediate ( 17b ) was only generated in hOAT and not in GABA-AT. Molecular docking studies and p K
a computational calculations highlighted the importance of chirality and the endocyclic double bond for inhibitory activity. The turnover mechanism of 10b was supported by mass spectrometric analysis of dissociable products and fluoride ion release experiments. Notably, the stopped-flow experiments were highly consistent with the proposed mechanism, suggesting a relatively slow hydrolysis rate for hOAT. The novel second-deprotonation mechanism of 10b contributes to its high potency and significantly enhanced selectivity for hOAT inhibition.- Published
- 2022
- Full Text
- View/download PDF
17. Inhibition of interferon-gamma-stimulated melanoma progression by targeting neuronal nitric oxide synthase (nNOS).
- Author
-
Tong S, Cinelli MA, El-Sayed NS, Huang H, Patel A, Silverman RB, and Yang S
- Subjects
- Animals, B7-H1 Antigen metabolism, Carcinogenesis metabolism, Cell Line, Tumor, Cell Survival drug effects, Humans, Interferon-alpha pharmacology, Melanoma pathology, Mice, Mice, Nude, Nitric Oxide Synthase Type I metabolism, STAT1 Transcription Factor metabolism, STAT3 Transcription Factor metabolism, Skin Neoplasms pathology, Treatment Outcome, Tumor Burden drug effects, Xenograft Model Antitumor Assays, Carcinogenesis chemically induced, Carcinogenesis drug effects, Disease Progression, Enzyme Inhibitors administration & dosage, Interferon-gamma administration & dosage, Melanoma drug therapy, Melanoma metabolism, Nitric Oxide Synthase Type I antagonists & inhibitors, Signal Transduction drug effects, Skin Neoplasms drug therapy, Skin Neoplasms metabolism
- Abstract
Interferon-gamma (IFN-γ) is shown to stimulate melanoma development and progression. However, the underlying mechanism has not been completely defined. Our study aimed to determine the role of neuronal nitric oxide synthase (nNOS)-mediated signaling in IFN-γ-stimulated melanoma progression and the anti-melanoma effects of novel nNOS inhibitors. Our study shows that IFN-γ markedly induced the expression levels of nNOS in melanoma cells associated with increased intracellular nitric oxide (NO) levels. Co-treatment with novel nNOS inhibitors effectively alleviated IFN-γ-activated STAT1/3. Further, reverse phase protein array (RPPA) analysis demonstrated that IFN-γ induced the expression of HIF1α, c-Myc, and programmed death-ligand 1 (PD-L1), in contrast to IFN-α. Blocking the nNOS-mediated signaling pathway using nNOS-selective inhibitors was shown to effectively diminish IFN-γ-induced PD-L1 expression in melanoma cells. Using a human melanoma xenograft mouse model, the in vivo studies revealed that IFN-γ increased tumor growth compared to control, which was inhibited by the co-administration of nNOS inhibitor MAC-3-190. Another nNOS inhibitor, HH044, was shown to effectively inhibit in vivo tumor growth and was associated with reduced PD-L1 expression levels in melanoma xenografts. Our study demonstrates the important role of nNOS-mediated NO signaling in IFN-γ-stimulated melanoma progression. Targeting nNOS using highly selective small molecular inhibitors is a unique and effective strategy to improve melanoma treatment., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
18. Inactivators of Ornithine Aminotransferase for the Treatment of Hepatocellular Carcinoma.
- Author
-
Silverman RB
- Abstract
Hepatocellular carcinoma (HCC) is the second or third leading cause of cancer mortality worldwide (depending on which statistics are used), yet there is no effective treatment. Currently, there are nine FDA-approved drugs for HCC, five monoclonal antibodies and four tyrosine kinase inhibitors. Ornithine aminotransferase (OAT) has been validated as a target in preclinical studies, which demonstrates that it is a potential target to treat HCC. Currently, there are no OAT inactivators in clinical trials for HCC. This Innovation describes evidence to support inhibition of OAT as a novel approach for HCC tumor growth inhibition. After the mechanism of OAT is discussed, the origins of our involvement in OAT inactivation, based on our previous work on mechanism-based inactivation of GABA-AT, are described. Once it was demonstrated that OAT inactivation does lead to HCC tumor growth inhibition, new selective OAT inactivators were designed and their inactivation mechanisms were elucidated. A summary of these mechanistic studies is presented. Inactivators of OAT provide the potential for treatment of HCC, targeting the Wnt/β-catenin pathway., Competing Interests: The author declares no competing financial interest., (© 2021 American Chemical Society.)
- Published
- 2021
- Full Text
- View/download PDF
19. OV329, a novel highly potent γ-aminobutyric acid aminotransferase inactivator, induces pronounced anticonvulsant effects in the pentylenetetrazole seizure threshold test and in amygdala-kindled rats.
- Author
-
Feja M, Meller S, Deking LS, Kaczmarek E, During MJ, Silverman RB, and Gernert M
- Subjects
- Amygdala, Animals, Anticonvulsants adverse effects, Female, Humans, Pentylenetetrazole adverse effects, Rats, Rats, Wistar, Seizures chemically induced, Seizures drug therapy, Transaminases adverse effects, Vigabatrin adverse effects, gamma-Aminobutyric Acid pharmacology, Epilepsy drug therapy, Kindling, Neurologic physiology
- Abstract
Objective: An attractive target to interfere with epileptic brain hyperexcitability is the enhancement of γ-aminobutyric acidergic (GABAergic) inhibition by inactivation of the GABA-metabolizing enzyme GABA aminotransferase (GABA-AT). GABA-AT inactivators were designed to control seizures by raising brain GABA levels. OV329, a novel drug candidate for the treatment of epilepsy and addiction, has been shown in vitro to be substantially more potent as a GABA-AT inactivator than vigabatrin, an antiseizure drug approved as an add-on therapy for adult patients with refractory complex partial seizures and monotherapy for pediatric patients with infantile spasms. Thus, we hypothesized that OV329 should produce pronounced anticonvulsant effects in two different rat seizure models., Methods: We therefore examined the effects of OV329 (5, 20, and 40 mg/kg ip) on the seizure threshold of female Wistar Unilever rats, using the timed intravenous pentylenetetrazole (ivPTZ) seizure threshold model as a seizure test particularly sensitive to GABA-potentiating manipulations, and amygdala-kindled rats as a model of difficult-to-treat temporal lobe epilepsy., Results: GABA-AT inactivation by OV329 clearly increased the threshold of both ivPTZ-induced and amygdala-kindled seizures. OV329 further showed a 30-fold greater anticonvulsant potency on ivPTZ-induced myoclonic jerks and clonic seizures compared to vigabatrin investigated previously. Notably, all rats were responsive to OV329 in both seizure models., Significance: These results reveal an anticonvulsant profile of OV329 that appears to be superior in both potency and efficacy to vigabatrin and highlight OV329 as a highly promising candidate for the treatment of seizures and pharmacoresistant epilepsies., (© 2021 The Authors. Epilepsia published by Wiley Periodicals LLC on behalf of International League Against Epilepsy.)
- Published
- 2021
- Full Text
- View/download PDF
20. Correction to A Single Amino Acid Determines the Selectivity and Efficacy of Selective Negative Allosteric Modulators of Ca V 1.3 L-Type Calcium Channels.
- Author
-
Cooper G, Kang S, Perez-Rosello T, Guzman JN, Galtieri D, Xie Z, Kondapalli J, Mordell J, Silverman RB, and Surmeier DJ
- Published
- 2021
- Full Text
- View/download PDF
21. Turnover and Inactivation Mechanisms for ( S )-3-Amino-4,4-difluorocyclopent-1-enecarboxylic Acid, a Selective Mechanism-Based Inactivator of Human Ornithine Aminotransferase.
- Author
-
Shen S, Butrin A, Doubleday PF, Melani RD, Beaupre BA, Tavares MT, Ferreira GM, Kelleher NL, Moran GR, Liu D, and Silverman RB
- Subjects
- Humans, Molecular Structure, Ornithine-Oxo-Acid Transaminase chemistry, Ornithine-Oxo-Acid Transaminase metabolism
- Abstract
The inhibition of human ornithine δ-aminotransferase ( h OAT) is a potential therapeutic approach to treat hepatocellular carcinoma. In this work, ( S )-3-amino-4,4-difluorocyclopent-1-enecarboxylic acid (SS-1-148, 6 ) was identified as a potent mechanism-based inactivator of h OAT while showing excellent selectivity over other related aminotransferases (e.g., GABA-AT). An integrated mechanistic study was performed to investigate the turnover and inactivation mechanisms of 6 . A monofluorinated ketone ( M10 ) was identified as the primary metabolite of 6 in h OAT. By soaking h OAT holoenzyme crystals with 6 , a precursor to M10 was successfully captured. This gem -diamine intermediate, covalently bound to Lys292, observed for the first time in h OAT/ligand crystals, validates the turnover mechanism proposed for 6 . Co-crystallization yielded h OAT in complex with 6 and revealed a novel noncovalent inactivation mechanism in h OAT. Native protein mass spectrometry was utilized for the first time in a study of an aminotransferase inactivator to validate the noncovalent interactions between the ligand and the enzyme; a covalently bonded complex was also identified as a minor form observed in the denaturing intact protein mass spectrum. Spectral and stopped-flow kinetic experiments supported a lysine-assisted E2 fluoride ion elimination, which has never been observed experimentally in other studies of related aminotransferase inactivators. This elimination generated the second external aldimine directly from the initial external aldimine, rather than the typical E1cB elimination mechanism, forming a quinonoid transient state between the two external aldimines. The use of native protein mass spectrometry, X-ray crystallography employing both soaking and co-crystallization methods, and stopped-flow kinetics allowed for the detailed elucidation of unusual turnover and inactivation pathways.
- Published
- 2021
- Full Text
- View/download PDF
22. Remarkable and Unexpected Mechanism for ( S )-3-Amino-4-(difluoromethylenyl)cyclohex-1-ene-1-carboxylic Acid as a Selective Inactivator of Human Ornithine Aminotransferase.
- Author
-
Zhu W, Doubleday PF, Butrin A, Weerawarna PM, Melani RD, Catlin DS, Dwight TA, Liu D, Kelleher NL, and Silverman RB
- Subjects
- Enzyme Inhibitors chemical synthesis, Enzyme Inhibitors chemistry, Humans, Kinetics, Mass Spectrometry, Models, Molecular, Molecular Structure, Ornithine-Oxo-Acid Transaminase metabolism, Enzyme Inhibitors pharmacology, Ornithine-Oxo-Acid Transaminase antagonists & inhibitors
- Abstract
Human ornithine aminotransferase ( h OAT) is a pyridoxal 5'-phosphate (PLP)-dependent enzyme that was recently found to play an important role in the metabolic reprogramming of hepatocellular carcinoma (HCC) via the proline and glutamine metabolic pathways. The selective inhibition of h OAT by compound 10 exhibited potent in vivo antitumor activity. Inspired by the discovery of the aminotransferase inactivator (1 S ,3 S )-3-amino-4-(difluoromethylene)cyclopentane-1-carboxylic acid ( 5 ), we rationally designed, synthesized, and evaluated a series of six-membered-ring analogs. Among them, 14 was identified as a new selective h OAT inactivator, which demonstrated a potency 22× greater than that of 10 . Three different types of protein mass spectrometry approaches and two crystallographic approaches were employed to identify the structure of h OAT- 14 and the formation of a remarkable final adduct ( 32' ) in the active site. These spectral studies reveal an enzyme complex heretofore not observed in a PLP-dependent enzyme, which has covalent bonds to two nearby residues. Crystal soaking experiments and molecular dynamics simulations were carried out to identify the structure of the active-site intermediate 27' and elucidate the order of the two covalent bonds that formed, leading to 32' . The initial covalent reaction of the activated warhead occurs with *Thr322 from the second subunit, followed by a subsequent nucleophilic attack by the catalytic residue Lys292. The turnover mechanism of 14 by h OAT was supported by a mass spectrometric analysis of metabolites and fluoride ion release experiments. This novel mechanism for h OAT with 14 will contribute to the further rational design of selective inactivators and an understanding of potential inactivation mechanisms by aminotransferases.
- Published
- 2021
- Full Text
- View/download PDF
23. Theoretical and Mechanistic Validation of Global Kinetic Parameters of the Inactivation of GABA Aminotransferase by OV329 and CPP-115.
- Author
-
Weerawarna PM, Moschitto MJ, and Silverman RB
- Subjects
- 4-Aminobutyrate Transaminase antagonists & inhibitors, Catalysis, Kinetics, Molecular Dynamics Simulation, Proline pharmacology, Quantum Theory, Reproducibility of Results, 4-Aminobutyrate Transaminase metabolism, Proline analogs & derivatives
- Abstract
(( S )-3-Amino-(difluoromethylenyl)cyclopent-1-ene-1-carboxylic acid (OV329) is a recently discovered inactivator of γ-aminobutyric acid aminotransferase (GABA-AT), which has 10 times better inactivation efficiency than its predecessor, CPP-115, despite the only structural difference being an endocyclic double bond in OV329. Both compounds are mechanism-based enzyme inactivators (MBEIs), which inactivate GABA-AT by a similar mechanism. Here, a combination of a variety of computational chemistry tools and experimental methods, including quantum mechanical (QM) calculations, molecular dynamic simulations, progress curve analysis, and deuterium kinetic isotope effect (KIE) experiments, are utilized to comprehensively study the mechanism of inactivation of GABA-AT by CPP-115 and OV329 and account for their experimentally obtained global kinetic parameters k
inact and KI . Our first key finding is that the rate-limiting step of the inactivation mechanism is the deprotonation step, and according to QM calculations and the KIE experiments, kinact accurately represents the enhancement of the rate-limiting step for the given mechanism. Second, the present study shows that the widely used simple QM models do not accurately represent the geometric criteria that are present in the enzyme for the deprotonation step. In contrast, QM cluster models successfully represent both the ground state destabilization and the transition state stabilization, as revealed by natural bond orbital analysis. Furthermore, the globally derived KI values for both of the inactivators represent the inhibitor constants for the initial binding complexes ( Kd ) and indicate the inactivator competition with the substrate according to progress curve analysis and the observed binding isotope effect. The configurational entropy loss accounts for the difference in KI values between the inactivators. The approach we describe in this work can be employed to determine the validity of globally derived parameters in the process of MBEI optimization for given inactivation mechanisms.- Published
- 2021
- Full Text
- View/download PDF
24. Improving mitochondria and ER stability helps eliminate upper motor neuron degeneration that occurs due to mSOD1 toxicity and TDP-43 pathology.
- Author
-
Genç B, Gautam M, Gözütok Ö, Dervishi I, Sanchez S, Goshu GM, Koçak N, Xie E, Silverman RB, and Özdinler PH
- Subjects
- Animals, Endoplasmic Reticulum metabolism, Female, Male, Mice, Mice, Inbred C57BL, Mitochondria metabolism, Motor Neuron Disease metabolism, Proteostasis Deficiencies metabolism, Rotarod Performance Test, TDP-43 Proteinopathies metabolism, Endoplasmic Reticulum pathology, Mitochondria pathology, Motor Neuron Disease pathology, Proteostasis Deficiencies pathology, Superoxide Dismutase-1 metabolism, TDP-43 Proteinopathies pathology
- Abstract
Background: Upper motor neurons (UMNs) are a key component of motor neuron circuitry. Their degeneration is a hallmark for diseases, such as hereditary spastic paraplegia (HSP), primary lateral sclerosis (PLS), and amyotrophic lateral sclerosis (ALS). Currently there are no preclinical assays investigating cellular responses of UMNs to compound treatment, even for diseases of the UMNs. The basis of UMN vulnerability is not fully understood, and no compound has yet been identified to improve the health of diseased UMNs: two major roadblocks for building effective treatment strategies., Methods: Novel UMN reporter models, in which UMNs that are diseased because of misfolded superoxide dismutase protein (mSOD1) toxicity and TDP-43 pathology are labeled with eGFP expression, allow direct assessment of UMN response to compound treatment. Electron microscopy reveals very precise aspects of endoplasmic reticulum (ER) and mitochondrial damage. Administration of NU-9, a compound initially identified based on its ability to reduce mSOD1 toxicity, has profound impact on improving the health and stability of UMNs, as identified by detailed cellular and ultrastructural analyses., Results: Problems with mitochondria and ER are conserved in diseased UMNs among different species. NU-9 has drug-like pharmacokinetic properties. It lacks toxicity and crosses the blood brain barrier. NU-9 improves the structural integrity of mitochondria and ER, reduces levels of mSOD1, stabilizes degenerating UMN apical dendrites, improves motor behavior measured by the hanging wire test, and eliminates ongoing degeneration of UMNs that become diseased both because of mSOD1 toxicity and TDP-43 pathology, two distinct and important overarching causes of motor neuron degeneration., Conclusions: Mechanism-focused and cell-based drug discovery approaches not only addressed key cellular defects responsible for UMN loss, but also identified NU-9, the first compound to improve the health of diseased UMNs, neurons that degenerate in ALS, HSP, PLS, and ALS/FTLD patients., (© 2021 The Authors. Clinical and Translational Medicine published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics.)
- Published
- 2021
- Full Text
- View/download PDF
25. Structural and Kinetic Analyses Reveal the Dual Inhibition Modes of Ornithine Aminotransferase by (1 S ,3 S )-3-Amino-4-(hexafluoropropan-2-ylidenyl)-cyclopentane-1-carboxylic Acid (BCF 3 ).
- Author
-
Butrin A, Beaupre BA, Kadamandla N, Zhao P, Shen S, Silverman RB, Moran GR, and Liu D
- Subjects
- Animals, Cell Line, Tumor, Crystallography, X-Ray, Enzyme Inhibitors pharmacology, Kinetics, Magnetic Resonance Spectroscopy methods, Mass Spectrometry methods, Mice, Molecular Structure, Xenograft Model Antitumor Assays, Carcinoma, Hepatocellular pathology, Enzyme Inhibitors chemistry, Liver Neoplasms pathology, Ornithine-Oxo-Acid Transaminase antagonists & inhibitors
- Abstract
Hepatocellular carcinoma (HCC) is the most common form of liver cancer and the leading cause of death among people with cirrhosis. HCC is typically diagnosed in advanced stages when tumors are resistant to both radio- and chemotherapy. Human ornithine aminotransferase ( h OAT) is a pyridoxal-5'-phosphate (PLP)-dependent enzyme involved in glutamine and proline metabolism. Because h OAT is overexpressed in HCC cells and a contributing factor for the uncontrolled cellular division that propagates malignant tumors (Ueno et al. J. Hepatol. 2014, 61, 1080-1087), it is a potential drug target for the treatment of HCC. (1 S ,3 S )-3-Amino-4-(hexafluoropropan-2-ylidenyl)-cyclopentane-1-carboxylic acid (BCF
3 ) has been shown in animal models to slow the progression of HCC by acting as a selective and potent mechanism-based inactivator of OAT (Zigmond et al. ACS Med. Chem. Lett. 2015, 6, 840-844). Previous studies have shown that the BCF3 - h OAT reaction has a bifurcation in which only 8% of the inhibitor inactivates the enzyme while the remaining 92% ultimately acts as a substrate and undergoes hydrolysis to regenerate the active PLP form of the enzyme. In this manuscript, the rate-limiting step of the inactivation mechanism was determined by stopped-flow spectrophotometry and time-dependent19 F NMR experiments to be the decay of a long-lived external aldimine species. A crystal structure of this transient complex revealed both the structural basis for fractional irreversible inhibition and the principal mode of inhibition of h OAT by BCF3 , which is to trap the enzyme in this transient but quasi-stable external aldimine form.- Published
- 2021
- Full Text
- View/download PDF
26. Pregabalin Treatment does not Affect Amyloid Pathology in 5XFAD Mice.
- Author
-
Sadleir KR, Popovoic J, Zhu W, Reidel CT, Do H, Silverman RB, and Vassar R
- Subjects
- Animals, Disease Models, Animal, Hippocampus metabolism, Humans, Mice, Mice, Transgenic, Neurites pathology, Alzheimer Disease drug therapy, Alzheimer Disease pathology, Amyloid Precursor Protein Secretases metabolism, Plaque, Amyloid pathology, Pregabalin therapeutic use
- Abstract
Background: Calcium dysregulation has been proposed to play a causative role in the development of Alzheimer's disease pathology. Pregabalin is a compound already approved for human use, marketed as the prescription drug Lyrica. It binds the α2-δ subunit of P/Q-type voltagegated calcium channels, lowering calcium influx and providing effective treatment for epilepsy and neuropathic pain., Objective: We hypothesize that increased resting calcium in neuronal processes near amyloid plaques plays a role in the development of neuritic dystrophies and further progression of amyloid pathology., Methods: 5XFAD mice were treated orally for 12 weeks with pregabalin, then immunoblotting and immunofluorescent imaging were used to quantify neuritic dystrophy and amyloid deposition in pregabalin compared to placebo-treated mice., Results: The treatment did not decrease markers of neuritic dystrophy or amyloid deposition. The image analysis of neuritic dystrophy on a plaque-by-plaque basis showed a small non-significant increase in the relative proportion of LAMP1 to Aβ42 in plaques with areas of 50-450 μm2 in the cortex of pregabalin-treated mice. In addition, there was a statistically significant positive correlation between the measured cerebral concentration of pregabalin and the relative levels of BACE1 and Aβ in the cortex. This relationship was not observed in the hippocampus, and there was no increase in average Aβ levels in pregabalin treated mice compared to placebo. We confirmed previous findings that smaller amyloid plaques are associated with a greater degree of neuritic dystrophy., Conclusion: Pregabalin may have an effect on Aβ that merits further investigation, but our study does not suggest that pregabalin contributes substantially to amyloid pathology., (Copyright© Bentham Science Publishers; For any queries, please email at epub@benthamscience.net.)
- Published
- 2021
- Full Text
- View/download PDF
27. Physiological involvement of presynaptic L-type voltage-dependent calcium channels in GABA release of cerebellar molecular layer interneurons.
- Author
-
Rey S, Maton G, Satake S, Llano I, Kang S, Surmeier DJ, Silverman RB, and Collin T
- Subjects
- Animals, Cerebellum cytology, Female, Male, Organ Culture Techniques, Rats, Rats, Sprague-Dawley, Calcium Channels, L-Type physiology, Cerebellum physiology, Interneurons physiology, Presynaptic Terminals physiology, gamma-Aminobutyric Acid metabolism
- Abstract
While high threshold voltage-dependent Ca
2+ channels (VDCCs) of the N and P/Q families are crucial for evoked neurotransmitter release in the mammalian CNS, it remains unclear to what extent L-type Ca2+ channels (LTCCs), which have been mainly considered as acting at postsynaptic sites, participate in the control of transmitter release. Here, we investigate the possible role of LTCCs in regulating GABA release by cerebellar molecular layer interneurons (MLIs) from rats. We found that BayK8644 (BayK) markedly increases mIPSC frequency in MLIs and Purkinje cells (PCs), suggesting that LTCCs are expressed presynaptically. Furthermore, we observed (1) a potentiation of evoked IPSCs in the presence of BayK, (2) an inhibition of evoked IPSCs in the presence of the LTCC-specific inhibitor Compound 8 (Cp8), and (3) a strong reduction of mIPSC frequency by Cp8. BayK effects are reduced by dantrolene, suggesting that ryanodine receptors act in synergy with LTCCs. Finally, BayK enhances presynaptic AP-evoked Ca2+ transients and increases the frequency of spontaneous axonal Ca2+ transients observed in TTX. Taken together, our data demonstrate that LTCCs are of primary importance in regulating GABA release by MLIs., (© 2020 International Society for Neurochemistry.)- Published
- 2020
- Full Text
- View/download PDF
28. A Single Amino Acid Determines the Selectivity and Efficacy of Selective Negative Allosteric Modulators of Ca V 1.3 L-Type Calcium Channels.
- Author
-
Cooper G, Kang S, Perez-Rosello T, Guzman JN, Galtieri D, Xie Z, Kondapalli J, Mordell J, Silverman RB, and Surmeier DJ
- Subjects
- Allosteric Regulation, Allosteric Site, Animals, Calcium metabolism, Calcium Channels, L-Type chemistry, Calcium Channels, L-Type genetics, Dopaminergic Neurons drug effects, HEK293 Cells, Humans, Mice, Molecular Docking Simulation, Mutagenesis, Site-Directed, Mutation, Protein Binding, Rabbits, Rats, Calcium Channel Blockers metabolism, Calcium Channels, L-Type metabolism, Pyrimidinones metabolism
- Abstract
Ca
2+ channels with a CaV 1.3 pore-forming α1 subunit have been implicated in both neurodegenerative and neuropsychiatric disorders, motivating the development of selective and potent inhibitors of CaV 1.3 versus CaV 1.2 channels, the calcium channels implicated in hypertensive disorders. We have previously identified pyrimidine-2,4,6-triones (PYTs) that preferentially inhibit CaV 1.3 channels, but the structural determinants of their interaction with the channel have not been identified, impeding their development into drugs. By a combination of biochemical, computational, and molecular biological approaches, it was found that PYTs bind to the dihydropyridine (DHP) binding pocket of the CaV 1.3 subunit, establishing them as negative allosteric modulators of channel gating. Site-directed mutagenesis, based on homology models of CaV 1.3 and CaV 1.2 channels, revealed that a single amino acid residue within the DHP binding pocket (M1078) is responsible for the selectivity of PYTs for CaV 1.3 over CaV 1.2. In addition to providing direction for chemical optimization, these results suggest that, like dihydropyridines, PYTs have pharmacological features that could make them of broad clinical utility.- Published
- 2020
- Full Text
- View/download PDF
29. (S)-4-Amino-5-phenoxypentanoate designed as a potential selective agonist of the bacterial transcription factor GabR.
- Author
-
Catlin DS, Reidl CT, Trzupek TR, Silverman RB, Cannon BL, Becker DP, and Liu D
- Subjects
- Bacillus subtilis genetics, Bacillus subtilis metabolism, Bacterial Proteins genetics, Bacterial Proteins metabolism, Crystallography, X-Ray, Operon, Protein Domains, Transcription Factors genetics, Transcription Factors metabolism, Transcription, Genetic, Bacillus subtilis chemistry, Bacterial Proteins agonists, Bacterial Proteins chemistry, Transcription Factors agonists, Transcription Factors chemistry, Valerates chemistry
- Abstract
Addressing molecular recognition in the context of evolution requires pursuing new molecular targets to enable the development of agonists or antagonists with new mechanisms of action. Disruption of transcriptional regulation through targeting transcription factors that regulate the expression of key enzymes in bacterial metabolism may provide a promising method for controlling the bacterial metabolic pathways. To this end, we have selectively targeted a bacterial transcription regulator through the design and synthesis of a series of γ-aminobutyric acid (GABA) derivatives, including (S)-4-amino-5-phenoxypentanoate (4-phenoxymethyl-GABA), which are based on docking insights gained from a previously-solved crystal structure of GabR from Bacillus subtilis. This target was selected because GabR strictly controls GABA metabolism by regulating the transcription of the gabT/D operon. These GabR transcription modulators are selective for the bacterial transcription factor GabR and are unable to bind to structural homologs of GabR due to distinct steric constraints. We have obtained a crystal structure of 4-phenoxymethyl-GABA bound as an external aldimine with PLP in the effector binding site of GabR, which suggests that this compound is capable of binding and reacting in the same manner as the native effector ligand. Inhibition assays demonstrate high selectivity of 4-phenoxymethyl-GABA for bacterial GabR versus several selected eukaryotic enzymes. Single-molecule fluorescence resonance energy transfer (smFRET) experiments reveal a ligand-induced DNA distortion that is very similar to that of the native effector GABA, suggesting that the compound functions as a potential selective agonist of GabR., (© 2020 The Protein Society.)
- Published
- 2020
- Full Text
- View/download PDF
30. First Contact: 7-Phenyl-2-Aminoquinolines, Potent and Selective Neuronal Nitric Oxide Synthase Inhibitors That Target an Isoform-Specific Aspartate.
- Author
-
Cinelli MA, Reidl CT, Li H, Chreifi G, Poulos TL, and Silverman RB
- Subjects
- Aminoquinolines chemical synthesis, Aminoquinolines metabolism, Aminoquinolines pharmacokinetics, Animals, Blood-Brain Barrier metabolism, Catalytic Domain, Crystallography, X-Ray, Enzyme Assays, Enzyme Inhibitors chemical synthesis, Enzyme Inhibitors metabolism, Enzyme Inhibitors pharmacokinetics, Humans, Isoenzymes antagonists & inhibitors, Isoenzymes chemistry, Isoenzymes genetics, Isoenzymes metabolism, Microsomes, Liver metabolism, Molecular Structure, Mutagenesis, Site-Directed, Mutation, Nitric Oxide Synthase Type I chemistry, Nitric Oxide Synthase Type I genetics, Nitric Oxide Synthase Type I metabolism, Permeability, Protein Binding, Rats, Structure-Activity Relationship, Aminoquinolines pharmacology, Aspartic Acid chemistry, Enzyme Inhibitors pharmacology, Nitric Oxide Synthase Type I antagonists & inhibitors
- Abstract
Inhibition of neuronal nitric oxide synthase (nNOS), an enzyme implicated in neurodegenerative disorders, is an attractive strategy for treating or preventing these diseases. We previously developed several classes of 2-aminoquinoline-based nNOS inhibitors, but these compounds had drawbacks including off-target promiscuity, low activity against human nNOS, and only modest selectivity for nNOS over related enzymes. In this study, we synthesized new nNOS inhibitors based on 7-phenyl-2-aminoquinoline and assayed them against rat and human nNOS, human eNOS, and murine and (in some cases) human iNOS. Compounds with a meta -relationship between the aminoquinoline and a positively charged tail moiety were potent and had up to nearly 900-fold selectivity for human nNOS over human eNOS. X-ray crystallography indicates that the amino groups of some compounds occupy a water-filled pocket surrounding an nNOS-specific aspartate residue (absent in eNOS). This interaction was confirmed by mutagenesis studies, making 7-phenyl-2-aminoquinolines the first aminoquinolines to interact with this residue.
- Published
- 2020
- Full Text
- View/download PDF
31. A Remarkable Difference That One Fluorine Atom Confers on the Mechanisms of Inactivation of Human Ornithine Aminotransferase by Two Cyclohexene Analogues of γ-Aminobutyric Acid.
- Author
-
Zhu W, Doubleday PF, Catlin DS, Weerawarna PM, Butrin A, Shen S, Wawrzak Z, Kelleher NL, Liu D, and Silverman RB
- Subjects
- Cyclohexanecarboxylic Acids chemical synthesis, Cyclohexanecarboxylic Acids metabolism, Cyclohexylamines chemical synthesis, Cyclohexylamines metabolism, Enzyme Inhibitors chemical synthesis, Enzyme Inhibitors metabolism, Humans, Hydrocarbons, Fluorinated chemical synthesis, Hydrocarbons, Fluorinated metabolism, Models, Chemical, Molecular Docking Simulation, Molecular Dynamics Simulation, Molecular Structure, Ornithine-Oxo-Acid Transaminase chemistry, Ornithine-Oxo-Acid Transaminase metabolism, Protein Binding, Pyridoxal Phosphate chemistry, gamma-Aminobutyric Acid analogs & derivatives, Cyclohexanecarboxylic Acids chemistry, Cyclohexylamines chemistry, Enzyme Inhibitors chemistry, Hydrocarbons, Fluorinated chemistry, Ornithine-Oxo-Acid Transaminase antagonists & inhibitors
- Abstract
Human ornithine aminotransferase ( h OAT), a pyridoxal 5'-phosphate-dependent enzyme, plays a critical role in the progression of hepatocellular carcinoma (HCC). Pharmacological selective inhibition of h OAT has been shown to be a potential therapeutic approach for HCC. Inspired by the discovery of the nonselective aminotransferase inactivator (1 R ,3 S ,4 S )-3-amino-4-fluoro cyclopentane-1-carboxylic acid ( 1 ), in this work, we rationally designed, synthesized, and evaluated a novel series of fluorine-substituted cyclohexene analogues, thereby identifying 8 and 9 as novel selective h OAT time-dependent inhibitors. Intact protein mass spectrometry and protein crystallography demonstrated 8 and 9 as covalent inhibitors of h OAT, which exhibit two distinct inactivation mechanisms resulting from the difference of a single fluorine atom. Interestingly, they share a similar turnover mechanism, according to the mass spectrometry-based analysis of metabolites and fluoride ion release experiments. Molecular dynamics (MD) simulations and electrostatic potential (ESP) charge calculations were conducted, which elucidated the significant influence of the one-fluorine difference on the corresponding intermediates, leading to two totally different inactivation pathways. The novel addition-aromatization inactivation mechanism for 9 contributes to its significantly enhanced potency, along with excellent selectivity over other aminotransferases.
- Published
- 2020
- Full Text
- View/download PDF
32. Mechanism-Based Design of 3-Amino-4-Halocyclopentenecarboxylic Acids as Inactivators of GABA Aminotransferase.
- Author
-
Shen S, Doubleday PF, Weerawarna PM, Zhu W, Kelleher NL, and Silverman RB
- Abstract
Aminotransferases are pyridoxal 5'-phosphate-dependent enzymes that catalyze reversible transamination reactions between an amino acid and an α-keto acid, playing a critical role in cellular nitrogen metabolism. It is evident that γ-aminobutyric acid aminotransferase (GABA-AT), which balances the levels of inhibitory and excitatory neurotransmitters, has emerged as a promising therapeutic target for epilepsy and cocaine addiction based on mechanism-based inactivators (MBIs). In this work, we established an integrated approach using computational simulation, organic synthesis, biochemical evaluation, and mass spectrometry to facilitate our design and mechanistic studies of MBIs, which led to the identification of a new cyclopentene-based analogue ( 6a ), 25-times more efficient as an inactivator of GABA-AT compared to the parent compound (1 R ,3 S ,4 S )-3-amino-4-fluorocyclopentane carboxylic acid ( FCP , 4 )., Competing Interests: The authors declare no competing financial interest.
- Published
- 2020
- Full Text
- View/download PDF
33. Inducible nitric oxide synthase: Regulation, structure, and inhibition.
- Author
-
Cinelli MA, Do HT, Miley GP, and Silverman RB
- Subjects
- Animals, Disease, Enzyme Inhibitors pharmacology, Humans, Models, Biological, Nitric Oxide biosynthesis, Nitric Oxide Synthase Type II genetics, Signal Transduction drug effects, Nitric Oxide Synthase Type II antagonists & inhibitors, Nitric Oxide Synthase Type II chemistry
- Abstract
A considerable number of human diseases have an inflammatory component, and a key mediator of immune activation and inflammation is inducible nitric oxide synthase (iNOS), which produces nitric oxide (NO) from l-arginine. Overexpressed or dysregulated iNOS has been implicated in numerous pathologies including sepsis, cancer, neurodegeneration, and various types of pain. Extensive knowledge has been accumulated about the roles iNOS plays in different tissues and organs. Additionally, X-ray crystal and cryogenic electron microscopy structures have shed new insights on the structure and regulation of this enzyme. Many potent iNOS inhibitors with high selectivity over related NOS isoforms, neuronal NOS, and endothelial NOS, have been discovered, and these drugs have shown promise in animal models of endotoxemia, inflammatory and neuropathic pain, arthritis, and other disorders. A major issue in iNOS inhibitor development is that promising results in animal studies have not translated to humans; there are no iNOS inhibitors approved for human use. In addition to assay limitations, both the dual modalities of iNOS and NO in disease states (ie, protective vs harmful effects) and the different roles and localizations of NOS isoforms create challenges for therapeutic intervention. This review summarizes the structure, function, and regulation of iNOS, with focus on the development of iNOS inhibitors (historical and recent). A better understanding of iNOS' complex functions is necessary before specific drug candidates can be identified for classical indications such as sepsis, heart failure, and pain; however, newer promising indications for iNOS inhibition, such as depression, neurodegenerative disorders, and epilepsy, have been discovered., (© 2019 Wiley Periodicals, Inc.)
- Published
- 2020
- Full Text
- View/download PDF
34. A modulator of wild-type glucocerebrosidase improves pathogenic phenotypes in dopaminergic neuronal models of Parkinson's disease.
- Author
-
Burbulla LF, Jeon S, Zheng J, Song P, Silverman RB, and Krainc D
- Subjects
- Animals, Brain metabolism, Cell Differentiation physiology, Dopaminergic Neurons metabolism, Glucosylceramidase genetics, Heterozygote, Humans, Induced Pluripotent Stem Cells metabolism, Mesencephalon metabolism, Mice, Neurons metabolism, Parkinson Disease genetics, Protein Binding, Spectroscopy, Near-Infrared, Tandem Mass Spectrometry, Glucosylceramidase metabolism, Parkinson Disease metabolism
- Abstract
Mutations in the GBA1 gene encoding the lysosomal enzyme β-glucocerebrosidase (GCase) represent the most common risk factor for Parkinson's disease (PD). GCase has been identified as a potential therapeutic target for PD and current efforts are focused on chemical chaperones to translocate mutant GCase into lysosomes. However, for several GBA1 -linked forms of PD and PD associated with mutations in LRRK2 , DJ-1 , and PARKIN , activating wild-type GCase represents an alternative approach. We developed a new small-molecule modulator of GCase called S-181 that increased wild-type GCase activity in iPSC-derived dopaminergic neurons from sporadic PD patients, as well as patients carrying the 84GG mutation in GBA1 , or mutations in LRRK2 , DJ-1 , or PARKIN who had decreased GCase activity. S-181 treatment of these PD iPSC-derived dopaminergic neurons partially restored lysosomal function and lowered accumulation of oxidized dopamine, glucosylceramide and α-synuclein. Moreover, S-181 treatment of mice heterozygous for the D409V GBA1 mutation ( Gba1
D409V/+ ) resulted in activation of wild-type GCase and consequent reduction of GCase lipid substrates and α-synuclein in mouse brain tissue. Our findings point to activation of wild-type GCase by small-molecule modulators as a potential therapeutic approach for treating familial and sporadic forms of PD that exhibit decreased GCase activity., (Copyright © 2019 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)- Published
- 2019
- Full Text
- View/download PDF
35. Mechanism of Inactivation of Ornithine Aminotransferase by (1 S ,3 S )-3-Amino-4-(hexafluoropropan-2-ylidenyl)cyclopentane-1-carboxylic Acid.
- Author
-
Moschitto MJ, Doubleday PF, Catlin DS, Kelleher NL, Liu D, and Silverman RB
- Subjects
- Carboxylic Acids chemistry, Carboxylic Acids pharmacology, Carcinoma, Hepatocellular enzymology, Halogenation, Humans, Liver Neoplasms enzymology, Models, Molecular, Ornithine-Oxo-Acid Transaminase chemistry, Ornithine-Oxo-Acid Transaminase metabolism, Cyclopentanes chemistry, Cyclopentanes pharmacology, Enzyme Inhibitors chemistry, Enzyme Inhibitors pharmacology, Ornithine-Oxo-Acid Transaminase antagonists & inhibitors
- Abstract
The inhibition of ornithine aminotransferase (OAT), a pyridoxal 5'-phosphate-dependent enzyme, has been implicated as a treatment for hepatocellular carcinoma (HCC), the most common form of liver cancer, for which there is no effective treatment. From a previous evaluation of our aminotransferase inhibitors, (1 S ,3 S )-3-amino-4-(perfluoropropan-2-ylidene)cyclopentane-1-carboxylic acid hydrochloride ( 1 ) was found to be a selective and potent inactivator of human OAT ( h OAT), which inhibited the growth of HCC in athymic mice implanted with human-derived HCC, even at a dose of 0.1 mg/kg. Currently, investigational new drug (IND)-enabling studies with 1 are underway. The inactivation mechanism of 1 , however, has proved to be elusive. Here we propose three possible mechanisms, based on mechanisms of known aminotransferase inactivators: Michael addition, enamine addition, and fluoride ion elimination followed by conjugate addition. On the basis of crystallography and intact protein mass spectrometry, it was determined that 1 inactivates h OAT through fluoride ion elimination to an activated 1,1'-difluoroolefin, followed by conjugate addition and hydrolysis. This result was confirmed with additional studies, including the detection of the cofactor structure by mass spectrometry and through the identification of turnover metabolites. On the basis of this inactivation mechanism and to provide further evidence for the mechanism, analogues of 1 ( 19 , 20 ) were designed, synthesized, and demonstrated to have the predicted selective inactivation mechanism. These analogues highlight the importance of the trifluoromethyl group and provide a basis for future inactivator design.
- Published
- 2019
- Full Text
- View/download PDF
36. Correction to Conversion of Quinazoline Modulators from Inhibitors to Activators of β-Glucocerebrosidase.
- Author
-
Zheng J, Jeon S, Jiang W, Burbulla LF, Ysselstein D, Oevel K, Krainc D, and Silverman RB
- Published
- 2019
- Full Text
- View/download PDF
37. Optimization of Blood-Brain Barrier Permeability with Potent and Selective Human Neuronal Nitric Oxide Synthase Inhibitors Having a 2-Aminopyridine Scaffold.
- Author
-
Do HT, Li H, Chreifi G, Poulos TL, and Silverman RB
- Subjects
- Animals, Caco-2 Cells, Enzyme Inhibitors chemistry, Humans, Permeability drug effects, Rats, Aminopyridines chemistry, Blood-Brain Barrier, Enzyme Inhibitors pharmacology, Nitric Oxide Synthase Type I antagonists & inhibitors
- Abstract
Effective delivery of therapeutic drugs into the human brain is one of the most challenging tasks in central nervous system drug development because of the blood-brain barrier (BBB). To overcome the BBB, both passive permeability and efflux transporter liability of a compound must be addressed. Herein, we report our optimization related to BBB penetration of neuronal nitric oxide synthase (nNOS) inhibitors toward the development of new drugs for neurodegenerative diseases. Various approaches, including enhancing lipophilicity and rigidity of new inhibitors and modulating the p K
a of amino groups, have been employed. In addition to determining inhibitor potency and selectivity, crystal structures of most newly designed compounds complexed to various nitric oxide synthase isoforms have been determined. We have discovered a new analogue (21), which exhibits not only excellent potency ( Ki < 30 nM) in nNOS inhibition but also a significantly low P-glycoprotein and breast-cancer-resistant protein substrate liability as indicated by an efflux ratio of 0.8 in the Caco-2 bidirectional assay.- Published
- 2019
- Full Text
- View/download PDF
38. Conversion of Quinazoline Modulators from Inhibitors to Activators of β-Glucocerebrosidase.
- Author
-
Zheng J, Jeon S, Jiang W, Burbulla LF, Ysselstein D, Oevel K, Krainc D, and Silverman RB
- Subjects
- Dopaminergic Neurons drug effects, Enzyme Activators chemistry, Enzyme Activators therapeutic use, Enzyme Inhibitors chemistry, Enzyme Inhibitors therapeutic use, Gaucher Disease drug therapy, Humans, Methylation, Parkinson Disease drug therapy, Parkinson Disease pathology, Quinazolines chemistry, Quinazolines therapeutic use, Structure-Activity Relationship, Enzyme Activators pharmacology, Enzyme Inhibitors pharmacology, Glucosylceramidase antagonists & inhibitors, Quinazolines pharmacology
- Abstract
Gaucher's disease is a lysosomal disease caused by mutations in the β-glucocerebrosidase gene ( GBA1 and GCase) that have been also linked to increased risk of Parkinson's disease (PD) and Diffuse Lewy body dementia. Prior studies have suggested that mutant GCase protein undergoes misfolding and degradation, and therefore, stabilization of the mutant protein represents an important therapeutic strategy in synucleinopathies. In this work, we present a structure-activity relationship (SAR) study of quinazoline compounds that serve as inhibitors of GCase. Unexpectedly, we found that N-methylation of these inhibitors transformed them into GCase activators. A systematic SAR study further revealed that replacement of the key oxygen atom in the linker of the quinazoline derivative also contributed to the activity switch. PD patient-derived fibroblasts and dopaminergic midbrain neurons were treated with a selected compound (9q) that partially stabilized GCase and improved its activity. These results highlight a novel strategy for therapeutic development of noninhibitory GCase modulators in PD and related synucleinopathies.
- Published
- 2019
- Full Text
- View/download PDF
39. Correction to Improvement of Cell Permeability of Human Neuronal Nitric Oxide Synthase Inhibitors Using Potent and Selective 2-Aminopyridine-Based Scaffolds with a Fluorobenzene Linker.
- Author
-
Do HT, Wang HY, Li H, Chreifi G, Poulos TL, and Silverman RB
- Published
- 2019
- Full Text
- View/download PDF
40. Correction to Nitrile in the Hole: Discovery of a Small Auxiliary Pocket in Neuronal Nitric Oxide Synthase Leading to the Development of Potent and Selective 2-Aminoquinoline Inhibitors.
- Author
-
Cinelli MA, Li H, Chreifi G, Poulos TL, and Silverman RB
- Published
- 2019
- Full Text
- View/download PDF
41. Structural Basis for Isoform Selective Nitric Oxide Synthase Inhibition by Thiophene-2-carboximidamides.
- Author
-
Li H, Evenson RJ, Chreifi G, Silverman RB, and Poulos TL
- Subjects
- Amides chemistry, Animals, Enzyme Inhibitors chemistry, Humans, Nitric Oxide Synthase Type I chemistry, Protein Isoforms, Rats, X-Ray Diffraction, Amides pharmacology, Carboxylic Acids chemistry, Enzyme Inhibitors pharmacology, Nitric Oxide Synthase Type I antagonists & inhibitors, Protein Conformation drug effects, Thiophenes chemistry
- Abstract
The overproduction of nitric oxide in the brain by neuronal nitric oxide synthase (nNOS) is associated with a number of neurodegenerative diseases. Although inhibiting nNOS is an important therapeutic goal, it is important not to inhibit endothelial NOS (eNOS) because of the critical role played by eNOS in maintaining vascular tone. While it has been possible to develop nNOS selective aminopyridine inhibitors, many of the most potent and selective inhibitors exhibit poor bioavailability properties. Our group and others have turned to more biocompatible thiophene-2-carboximidamide (T2C) inhibitors as potential nNOS selective inhibitors. We have used crystallography and computational methods to better understand how and why two commercially developed T2C inhibitors exhibit selectivity for human nNOS over human eNOS. As with many of the aminopyridine inhibitors, a critical active site Asp residue in nNOS versus Asn in eNOS is largely responsible for controlling selectivity. We also present thermodynamic integration results to better understand the change in p K
a and thus the charge of inhibitors once bound to the active site. In addition, relative free energy calculations underscore the importance of enhanced electrostatic stabilization of inhibitors bound to the nNOS active site compared to eNOS.- Published
- 2018
- Full Text
- View/download PDF
42. Synthesis of ( S)-3-Amino-4-(difluoromethylenyl)-cyclopent-1-ene-1-carboxylic Acid (OV329), a Potent Inactivator of γ-Aminobutyric Acid Aminotransferase.
- Author
-
Moschitto MJ and Silverman RB
- Subjects
- Molecular Structure, Organometallic Compounds chemistry, Phenylacetates chemistry, Selenium Compounds chemistry, Stereoisomerism, Structure-Activity Relationship, 4-Aminobutyrate Transaminase antagonists & inhibitors, Cyclopentanes chemistry, Enzyme Inhibitors chemical synthesis, Transaminases antagonists & inhibitors
- Abstract
( S)-3-Amino-4-(difluoromethylenyl)cyclopent-1-ene-1-carboxylic acid (OV329, 1) is being developed for the treatment of epilepsy and addiction. The previous 14-step synthesis of OV329 was low yielding, involved an unselective α-elimination to form the cyclopentene, required the use of tert-butyllithium, and produced toxic selenium byproducts in the penultimate step. A new synthesis, which avoids the aforementioned issues, was carried out on large scale, reducing the step count from 14 to 9 steps and increasing the overall yield from 3.7% to 8.1%.
- Published
- 2018
- Full Text
- View/download PDF
43. β-Glucocerebrosidase Modulators Promote Dimerization of β-Glucocerebrosidase and Reveal an Allosteric Binding Site.
- Author
-
Zheng J, Chen L, Skinner OS, Ysselstein D, Remis J, Lansbury P, Skerlj R, Mrosek M, Heunisch U, Krapp S, Charrow J, Schwake M, Kelleher NL, Silverman RB, and Krainc D
- Subjects
- Crystallography, X-Ray, Fibroblasts metabolism, Glucosylceramidase genetics, HEK293 Cells, Humans, Mass Spectrometry, Models, Molecular, Molecular Structure, Mutation, Allosteric Site drug effects, Glucosylceramidase chemistry, Glucosylceramidase metabolism, Protein Multimerization drug effects
- Abstract
β-Glucocerebrosidase (GCase) mutations cause Gaucher's disease and are a high risk factor in Parkinson's disease. The implementation of a small molecule modulator is a strategy to restore proper folding and lysosome delivery of degradation-prone mutant GCase. Here, we present a potent quinazoline modulator, JZ-4109, which stabilizes wild-type and N370S mutant GCase and increases GCase abundance in patient-derived fibroblast cells. We then developed a covalent modification strategy using a lysine targeted inactivator (JZ-5029) for in vitro mechanistic studies. By using native top-down mass spectrometry, we located two potentially covalently modified lysines. We obtained the first crystal structure, at 2.2 Å resolution, of a GCase with a noniminosugar modulator covalently bound, and were able to identify the exact lysine residue modified (Lys346) and reveal an allosteric binding site. GCase dimerization was induced by our modulator binding, which was observed by native mass spectrometry, its crystal structure, and size exclusion chromatography with a multiangle light scattering detector. Finally, the dimer form was confirmed by negative staining transmission electron microscopy studies. Our newly discovered allosteric site and observed GCase dimerization provide a new mechanistic insight into GCase and its noniminosugar modulators and facilitate the rational design of novel GCase modulators for Gaucher's disease and Parkinson's disease.
- Published
- 2018
- Full Text
- View/download PDF
44. Total Synthesis of Tambromycin Enabled by Indole C-H Functionalization.
- Author
-
Miley GP, Rote JC, Silverman RB, Kelleher NL, and Thomson RJ
- Subjects
- Amination, Amino Acids, Molecular Structure, Tryptophan, Indoles chemistry
- Abstract
The total synthesis of tambromycin (1), a recently isolated tetrapeptide, is reported. This unusual natural product possesses a highly modified tryptophan-derived indole fragment fused to an α-methylserine-derived oxazoline ring, and a unique noncanonical amino acid residue named tambroline (11). A convergent synthesis of tambromycin was achieved by a 13-step route that leveraged recent developments in the field of C-H functionalization to prepare the complex indole fragment, as well as an efficient synthesis of tambroline that featured a diastereoselective amination of homoproline.
- Published
- 2018
- Full Text
- View/download PDF
45. Design and Mechanism of GABA Aminotransferase Inactivators. Treatments for Epilepsies and Addictions.
- Author
-
Silverman RB
- Subjects
- 4-Aminobutyrate Transaminase metabolism, Animals, Drug Design, Enzyme Inhibitors pharmacology, Humans, Models, Molecular, 4-Aminobutyrate Transaminase antagonists & inhibitors, Enzyme Inhibitors chemistry, Epilepsy drug therapy, Substance-Related Disorders drug therapy
- Abstract
When the brain concentration of the inhibitory neurotransmitter γ-aminobutyric acid (GABA) diminishes below a threshold level, the excess neuronal excitation can lead to convulsions. This imbalance in neurotransmission can be corrected by inhibition of the enzyme γ-aminobutyric acid aminotransferase (GABA-AT), which catalyzes the conversion of GABA to the excitatory neurotransmitter l-glutamic acid. It also has been found that raising GABA levels can antagonize the rapid elevation and release of dopamine in the nucleus accumbens, which is responsible for the reward response in addiction. Therefore, the design of new inhibitors of GABA-AT, which increases brain GABA levels, is an important approach to new treatments for epilepsy and addiction. This review summarizes findings over the last 40 or so years of mechanism-based inactivators (unreactive compounds that require the target enzyme to catalyze their conversion to the inactivating species, which inactivate the enzyme prior to their release) of GABA-AT with emphasis on their catalytic mechanisms of inactivation, presented according to organic chemical mechanism, with minimal pharmacology, except where important for activity in epilepsy and addiction. Patents, abstracts, and conference proceedings are not covered in this review. The inactivation mechanisms described here can be applied to the inactivations of a wide variety of unrelated enzymes.
- Published
- 2018
- Full Text
- View/download PDF
46. Design and Mechanism of (S)-3-Amino-4-(difluoromethylenyl)cyclopent-1-ene-1-carboxylic Acid, a Highly Potent γ-Aminobutyric Acid Aminotransferase Inactivator for the Treatment of Addiction.
- Author
-
Juncosa JI, Takaya K, Le HV, Moschitto MJ, Weerawarna PM, Mascarenhas R, Liu D, Dewey SL, and Silverman RB
- Subjects
- 4-Aminobutyrate Transaminase chemistry, 4-Aminobutyrate Transaminase metabolism, Animals, Brain drug effects, Brain metabolism, Catalytic Domain drug effects, Crystallography, X-Ray, Dopamine metabolism, Dopamine Antagonists chemistry, Dopamine Antagonists pharmacokinetics, Dopamine Antagonists pharmacology, Enzyme Inhibitors pharmacokinetics, Glucose metabolism, Humans, Male, Models, Molecular, Proline chemistry, Proline pharmacokinetics, Proline pharmacology, Rats, Rats, Sprague-Dawley, gamma-Aminobutyric Acid metabolism, 4-Aminobutyrate Transaminase antagonists & inhibitors, Drug Design, Enzyme Inhibitors chemistry, Enzyme Inhibitors pharmacology, Proline analogs & derivatives
- Abstract
γ-Aminobutyric acid (GABA) is the major inhibitory neurotransmitter in the central nervous system. Inhibition of GABA aminotransferase (GABA-AT), a pyridoxal 5'-phosphate (PLP)-dependent enzyme that degrades GABA, has been established as a possible strategy for the treatment of substance abuse. The raised GABA levels that occur as a consequence of this inhibition have been found to antagonize the rapid release of dopamine in the ventral striatum (nucleus accumbens) that follows an acute challenge by an addictive substance. In addition, increased GABA levels are also known to elicit an anticonvulsant effect in patients with epilepsy. We previously designed the mechanism-based inactivator (1S,3S)-3-amino-4-difluoromethylenyl-1-cyclopentanoic acid (2), now called CPP-115, that is 186 times more efficient in inactivating GABA-AT than vigabatrin, the only FDA-approved drug that is an inactivator of GABA-AT. CPP-115 was found to have high therapeutic potential for the treatment of cocaine addiction and for a variety of epilepsies, has successfully completed a Phase I safety clinical trial, and was found to be effective in the treatment of infantile spasms (West syndrome). Herein we report the design, using molecular dynamics simulations, synthesis, and biological evaluation of a new mechanism-based inactivator, (S)-3-amino-4-(difluoromethylenyl)cyclopent-1-ene-1-carboxylic acid (5), which was found to be almost 10 times more efficient as an inactivator of GABA-AT than CPP-115. We also present the unexpected crystal structure of 5 bound to GABA-AT, as well as computational analyses used to assist the structure elucidation process. Furthermore, 5 was found to have favorable pharmacokinetic properties and low off-target activities. In vivo studies in freely moving rats showed that 5 was dramatically superior to CPP-115 in suppressing the release of dopamine in the corpus striatum, which occurs subsequent to either an acute cocaine or nicotine challenge. Compound 5 also attenuated increased metabolic demands (neuronal glucose metabolism) in the hippocampus, a brain region that encodes spatial information concerning the environment in which an animal receives a reinforcing or aversive drug. This multidisciplinary computational design to preclinical efficacy approach should be applicable to the design and improvement of mechanism-based inhibitors of other enzymes whose crystal structures and inactivation mechanisms are known.
- Published
- 2018
- Full Text
- View/download PDF
47. Improvement of Cell Permeability of Human Neuronal Nitric Oxide Synthase Inhibitors Using Potent and Selective 2-Aminopyridine-Based Scaffolds with a Fluorobenzene Linker.
- Author
-
Do HT, Wang HY, Li H, Chreifi G, Poulos TL, and Silverman RB
- Subjects
- Aminopyridines chemical synthesis, Animals, Blood-Brain Barrier metabolism, Caco-2 Cells, Crystallography, X-Ray, Fluorobenzenes chemical synthesis, Humans, Mice, Nitric Oxide Synthase Type II antagonists & inhibitors, Nitric Oxide Synthase Type III antagonists & inhibitors, Rats, Structure-Activity Relationship, Swine, Theophylline pharmacology, Verapamil pharmacology, Aminopyridines pharmacology, Cell Membrane Permeability drug effects, Fluorobenzenes pharmacology, Nitric Oxide Synthase Type I antagonists & inhibitors
- Abstract
Inhibition of neuronal nitric oxide synthase (nNOS) is a promising therapeutic approach to treat neurodegenerative diseases. Recently, we have achieved considerable progress in improving the potency and isoform selectivity of human nNOS inhibitors bearing a 2-aminopyridine scaffold. However, these inhibitors still suffered from too low cell membrane permeability to enter into CNS drug development. We report herein our studies to improve permeability of nNOS inhibitors as measured by both PAMPA-BBB and Caco-2 assays. The most permeable compound (12) in this study still preserves excellent potency with human nNOS (K
i = 30 nM) and very high selectivity over other NOS isoforms, especially human eNOS (hnNOS/heNOS = 2799, the highest hnNOS/heNOS ratio we have obtained to date). X-ray crystallographic analysis reveals that 12 adopts a similar binding mode in both rat and human nNOS, in which the 2-aminopyridine and the fluorobenzene linker form crucial hydrogen bonds with glutamate and tyrosine residues, respectively.- Published
- 2017
- Full Text
- View/download PDF
48. Correction to Selective Targeting by a Mechanism-Based Inactivator against Pyridoxal 5'-Phosphate-Dependent Enzymes: Mechanisms of Inactivation and Alternative Turnover.
- Author
-
Mascarenhas R, Le HV, Clevenger KD, Lehrer HJ, Ringe D, Kelleher NL, Silverman RB, and Liu D
- Published
- 2017
- Full Text
- View/download PDF
49. Selective Targeting by a Mechanism-Based Inactivator against Pyridoxal 5'-Phosphate-Dependent Enzymes: Mechanisms of Inactivation and Alternative Turnover.
- Author
-
Mascarenhas R, Le HV, Clevenger KD, Lehrer HJ, Ringe D, Kelleher NL, Silverman RB, and Liu D
- Subjects
- 4-Aminobutyrate Transaminase chemistry, 4-Aminobutyrate Transaminase metabolism, Aspartate Aminotransferases chemistry, Aspartate Aminotransferases genetics, Aspartate Aminotransferases metabolism, Binding Sites, Catalytic Domain, Crystallography, X-Ray, Cycloleucine chemistry, Cycloleucine metabolism, Cycloleucine pharmacology, Databases, Chemical, Databases, Protein, Enzyme Inhibitors chemistry, Enzyme Inhibitors metabolism, Escherichia coli Proteins antagonists & inhibitors, Escherichia coli Proteins chemistry, Escherichia coli Proteins genetics, Escherichia coli Proteins metabolism, Humans, Ligands, Molecular Conformation, Ornithine-Oxo-Acid Transaminase chemistry, Ornithine-Oxo-Acid Transaminase genetics, Ornithine-Oxo-Acid Transaminase metabolism, Protein Conformation, Pyridoxal Phosphate chemistry, Pyridoxamine chemistry, Pyridoxamine metabolism, Recombinant Proteins chemistry, Recombinant Proteins metabolism, Structural Homology, Protein, Substrate Specificity, 4-Aminobutyrate Transaminase antagonists & inhibitors, Aspartate Aminotransferases antagonists & inhibitors, Cycloleucine analogs & derivatives, Enzyme Inhibitors pharmacology, Models, Molecular, Ornithine-Oxo-Acid Transaminase antagonists & inhibitors, Pyridoxal Phosphate metabolism
- Abstract
Potent mechanism-based inactivators can be rationally designed against pyridoxal 5'-phosphate (PLP)-dependent drug targets, such as ornithine aminotransferase (OAT) or γ-aminobutyric acid aminotransferase (GABA-AT). An important challenge, however, is the lack of selectivity toward other PLP-dependent, off-target enzymes, because of similarities in mechanisms of all PLP-dependent aminotransferase reactions. On the basis of complex crystal structures, we investigate the inactivation mechanism of OAT, a hepatocellular carcinoma target, by (1R,3S,4S)-3-amino-4-fluorocyclopentane-1-carboxylic acid (FCP), a known inactivator of GABA-AT. A crystal structure of OAT and FCP showed the formation of a ternary adduct. This adduct can be rationalized as occurring via an enamine mechanism of inactivation, similar to that reported for GABA-AT. However, the crystal structure of an off-target, PLP-dependent enzyme, aspartate aminotransferase (Asp-AT), in complex with FCP, along with the results of attempted inhibition assays, suggests that FCP is not an inactivator of Asp-AT, but rather an alternate substrate. Turnover of FCP by Asp-AT is also supported by high-resolution mass spectrometry. Amid existing difficulties in achieving selectivity of inactivation among a large number of PLP-dependent enzymes, the obtained results provide evidence that a desirable selectivity could be achieved, taking advantage of subtle structural and mechanistic differences between a drug-target enzyme and an off-target enzyme, despite their largely similar substrate binding sites and catalytic mechanisms.
- Published
- 2017
- Full Text
- View/download PDF
50. Hydrophilic, Potent, and Selective 7-Substituted 2-Aminoquinolines as Improved Human Neuronal Nitric Oxide Synthase Inhibitors.
- Author
-
Pensa AV, Cinelli MA, Li H, Chreifi G, Mukherjee P, Roman LJ, Martásek P, Poulos TL, and Silverman RB
- Subjects
- Aminoquinolines chemical synthesis, Animals, Caco-2 Cells, Cattle, Cell Membrane Permeability drug effects, Enzyme Assays, Histidine chemistry, Humans, Mice, Nitric Oxide Synthase Type I chemistry, Nitric Oxide Synthase Type II antagonists & inhibitors, Nitric Oxide Synthase Type III antagonists & inhibitors, Rats, Aminoquinolines pharmacology, Nitric Oxide Synthase Type I antagonists & inhibitors
- Abstract
Neuronal nitric oxide synthase (nNOS) is a target for development of antineurodegenerative agents. Most nNOS inhibitors mimic l-arginine and have poor bioavailability. 2-Aminoquinolines showed promise as bioavailable nNOS inhibitors but suffered from low human nNOS inhibition, low selectivity versus human eNOS, and significant binding to other CNS targets. We aimed to improve human nNOS potency and selectivity and reduce off-target binding by (a) truncating the original scaffold or (b) introducing a hydrophilic group to interrupt the lipophilic, promiscuous pharmacophore and promote interaction with human nNOS-specific His342. We synthesized both truncated and polar 2-aminoquinoline derivatives and assayed them against recombinant NOS enzymes. Although aniline and pyridine derivatives interact with His342, benzonitriles conferred the best rat and human nNOS inhibition. Both introduction of a hydrophobic substituent next to the cyano group and aminoquinoline methylation considerably improved isoform selectivity. Most importantly, these modifications preserved Caco-2 permeability and reduced off-target CNS binding.
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.