31 results on '"S, Valente"'
Search Results
2. Chemical, Structural, and Morphological Changes of a MoVTeNb Catalyst during Oxidative Dehydrogenation of Ethane
- Author
-
Héctor Armendáriz-Herrera, Paz del Angel, R. Quintana-Solórzano, Jaime S. Valente, Jose Nieto, A. Massó, and N. Nava
- Subjects
Ethane ,Materials science ,Ethylene ,Rietveld refinement ,Inorganic chemistry ,General Chemistry ,Oxidative Dehydrogenation ,Structural Changes ,Catalysis ,symbols.namesake ,chemistry.chemical_compound ,chemistry ,Phase (matter) ,symbols ,Mo-V-Te-Nb oxides Catalysts ,Mixed oxide ,Morphological Changes ,Thermal stability ,Dehydrogenation ,Raman spectroscopy - Abstract
MoVTeNb mixed oxide, a highly active and selective catalyst for the oxidative dehydrogenation of ethane to produce ethylene, exhibits the so-called M1 and M2 crystalline phases. The thermal stability of the MoVTeNb catalytic system was assessed under varying reaction conditions; to this end, the catalyst was exposed to several reaction temperatures spanning from 440 to 550 °C. Both the pristine and spent materials were analyzed by several characterization techniques. The catalyst was stable below 500 °C; a reaction temperature of ≥500 °C brings about the removal of tellurium from the intercalated framework channels of the M1 crystalline phase. Rietveld refinement of X-ray diffraction patterns and microscopy results showed that the tellurium loss causes the progressive partial destruction of the M1 phase, thus decreasing the number of active sites and forming a MoO2 crystalline phase, which is inactive for this reaction. Raman spectroscopy confirmed the MoO2 phase development as a function of reaction temperature. From highresolution transmission electron microscopy and energy-dispersive X-ray spectroscopy analyses it was noticed that tellurium departure occurs preferentially from the end sides of the needlelike M1 crystals, across the [001] plane. Detailed analysis of a solid deposited at the reactor outlet showrf that it consisted mainly of metallic tellurium, suggesting that the tellurium detachment occurs via reduction of Te4+ to Te0 due to a combination of reaction temperature and feed composition. Thus, in order to sustain the catalytic performance exhibited by MoVTeNb mixed oxide, hot spots along the reactor bed should be avoided or controlled, maintaining the catalytic bed temperature below 500 °C., This work was financially supported by the Instituto Mexicano del Petroleo.
- Published
- 2014
3. Method for Large-Scale Production of Multimetallic Layered Double Hydroxides: Formation Mechanism Discernment.
- Author
-
Jaime S. Valente, Manuel Sánchez-Cantú, Enrique Lima, and François Figueras
- Subjects
- *
LAYERED double hydroxides , *REACTION mechanisms (Chemistry) , *METALLIC oxides , *NITRATES , *HYDROGEN-ion concentration , *PRECIPITATION (Chemistry) , *RECRYSTALLIZATION (Metallurgy) , *X-ray diffraction - Abstract
A simple, economical, and environmentally friendly method for the production of layered double hydroxides (LDHs) is presented. The synthesis procedure is based on dispersing insoluble metal oxides, adjusting the pH by adding an optimum amount of metal nitrates, and dispersing and aging the product for a short time (6−8 h). The final product does not require washing, opposite to the traditional coprecipitation synthesis procedure. A dissolution−precipitation−recrystallization mechanism is proposed for the formation of LDHs, based on particle size measurements, XRD analyses, radial distribution functions, and 27Al MAS NMR studies. Solids were characterized by XRD, N2physisorption, TGA-DTA, SEM, and TEM, revealing that both LDHs and their calcination products have very similar properties to those prepared by conventional procedures. Pure LDH phase is obtained after 6−8 h; a large, uniform particle size that would usually require prolonged hydrothermal treatments is attained. Surface areas ranged from 32 to 93 m2g−1and from 140 to 230 m2g−1for fresh and calcined samples, respectively. This new method is intended to satisfy the growing demand of LDHs in large-scale applications as catalysts, SOxadsorbents, PVC additives, etc. [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
4. Proposed General Sol−Gel Method to Prepare Multimetallic Layered Double Hydroxides: Synthesis, Characterization, and Envisaged Application.
- Author
-
Julia Prince, Ascencion Montoya, Gerardo Ferrat, and Jaime S. Valente
- Published
- 2009
- Full Text
- View/download PDF
5. A Simple Environmentally Friendly Method to Prepare Versatile Hydrotalcite-like Compounds.
- Author
-
Jaime S. Valente, Manuel S. Cantu, and François Figueras
- Published
- 2008
- Full Text
- View/download PDF
6. SIRT3 Activation a Promise in Drug Development? New Insights into SIRT3 Biology and Its Implications on the Drug Discovery Process.
- Author
-
Lambona C, Zwergel C, Valente S, and Mai A
- Subjects
- Animals, Biology, Drug Development, Drug Discovery, Mammals metabolism, Sirtuin 3 drug effects, Sirtuin 3 metabolism, Sirtuins drug effects, Sirtuins metabolism
- Abstract
Sirtuins catalyze deacetylation of lysine residues with a NAD
+ -dependent mechanism. In mammals, the sirtuin family is composed of seven members, divided into four subclasses that differ in substrate specificity, subcellular localization, regulation, as well as interactions with other proteins, both within and outside the epigenetic field. Recently, much interest has been growing in SIRT3, which is mainly involved in regulating mitochondrial metabolism. Moreover, SIRT3 seems to be protective in diseases such as age-related, neurodegenerative, liver, kidney, heart, and metabolic ones, as well as in cancer. In most cases, activating SIRT3 could be a promising strategy to tackle these health problems. Here, we summarize the main biological functions, substrates, and interactors of SIRT3, as well as several molecules reported in the literature that are able to modulate SIRT3 activity. Among the activators, some derive from natural products, others from library screening, and others from the classical medicinal chemistry approach.- Published
- 2024
- Full Text
- View/download PDF
7. Novel 1,4-Dihydropyridines as Specific Binders and Activators of SIRT3 Impair Cell Viability and Clonogenicity and Downregulate Hypoxia-Induced Targets in Cancer Cells.
- Author
-
Zwergel C, Aventaggiato M, Garbo S, Di Bello E, Fassari B, Noce B, Castiello C, Lambona C, Barreca F, Rotili D, Fioravanti R, Schmalz T, Weyand M, Niedermeier A, Tripodi M, Colotti G, Steegborn C, Battistelli C, Tafani M, Valente S, and Mai A
- Subjects
- Humans, Cell Survival, Cell Line, Tumor, Hypoxia, Hypoxia-Inducible Factor 1, alpha Subunit, Sirtuin 3, Neoplasms
- Abstract
The mitochondrial SIRT3 modulates several biological pathways such as cancer, metabolism, and hypoxia-related diseases. Recently, we discovered new 1,4-dihydropyridines, compounds 2 and 3 , the latter being a SIRT3-specific activator. In the present work, a novel 2 - and 3 -related small series of compounds have been developed, with 3c displaying the strongest SIRT3 binding and activation, with a K
D of 29 μM and 387% of enzyme activation. Differently, 3d was the best in enhancing glutamate dehydrogenase activity and deacetylating K68- and K122-acMnSOD in triple-negative MDA-MB-231 breast cancer cells. Tested in CAL-62 thyroid cancer and MDA-MB-231 cells, 3d displayed the strongest time- and dose-dependent reduction of cell viability and clonogenicity at a single-digit micromolar level, along with cell death, in both normoxia and hypoxia conditions. Moreover, 3d downregulated not only hypoxia-induced factors, such as HIF-1α, EPAS-1, and CA-IX, but also epithelial-mesenchymal transition master regulators and extracellular matrix components such as SNAIL1, ZEB1, SLUG, COL1A2, MMP2, and MMP9, markedly hampering MDA-MB-231 cell migration.- Published
- 2023
- Full Text
- View/download PDF
8. Hydrophobic DES Based on Menthol and Natural Organic Acids for Use in Antifouling Marine Coatings.
- Author
-
Valente S, Oliveira F, Ferreira IJ, Paiva A, Sobral RG, Diniz MS, Gaudêncio SP, and Duarte ARC
- Abstract
Marine biofouling negatively impacts industries with off-shore infrastructures, such as naval, oil, and aquaculture. To date, there are no ideal sustainable, economic, and environmentally benign solutions to deal with this phenomenon. The advances achieved in green solvents, as well as its application in different industries, such as pharmaceutical and biotechnology, have promoted the emergence of deep eutectic systems (DES). These eutectic systems have applications in various fields and can be revolutionary in the marine-based industrial sector. In this study, the main objective was to investigate the potential use of hydrophobic DES (HDES) based on menthol and natural organic acids for their use as marine antifouling coatings. Our strategy encompassed the physicochemical characterization of different formulations, which allowed us to identify the most appropriate molar ratio and intermolecular interactions for HDES formations. The miscibility of the resulting HDES with the marine coating has been evaluated and proven to be successful. The Men/OL (1:1) system proved to be the most promising in terms of cost-production and thus was the one used in subsequent antifouling tests. The cytotoxicity of this HDES was evaluated using an in vitro cell model (HaCat cells) showing no significant toxicity. Furthermore, the application of this system incorporated into coatings that are used in marine structures was also studied using marine species ( Mytilus edulis mussels and Patella vulgata limpets) to evaluate both their antifouling and ecotoxicity effects. HDES Men/OL (1:1) incorporated in marine coatings was promising in reducing marine macrofouling and also proved to be effective at the level of microfouling without viability impairment of the tested marine species. It was revealed to be more efficient than using copper oxide, metallic copper, or ivermectin as antifouling agents. Biochemical assays performed on marine species showed that this HDES does not induce oxidative stress in the tested species. These results are a strong indication of the potential of this HDES to be sustainable and efficiently used in marine fouling control technologies., Competing Interests: The authors declare no competing financial interest., (© 2023 The Authors. Published by American Chemical Society.)
- Published
- 2023
- Full Text
- View/download PDF
9. METTL3 from Target Validation to the First Small-Molecule Inhibitors: A Medicinal Chemistry Journey.
- Author
-
Fiorentino F, Menna M, Rotili D, Valente S, and Mai A
- Subjects
- RNA, Messenger metabolism, Methylation, RNA, Chemistry, Pharmaceutical, Methyltransferases chemistry
- Abstract
RNA methylation is a critical mechanism for regulating the transcription and translation of specific sequences or for eliminating unnecessary sequences during RNA maturation. METTL3, an RNA methyltransferase that catalyzes the transfer of a methyl group to the N
6 -adenosine of RNA, is one of the key mediators of this process. METTL3 dysregulation may result in the emergence of a variety of diseases ranging from cancer to cardiovascular and neurological disorders beyond contributing to viral infections. Hence, the discovery of METTL3 inhibitors may assist in furthering the understanding of the biological roles of this enzyme, in addition to contributing to the development of novel therapeutics. Through this work, we will examine the existing correlations between METTL3 and diseases. We will also analyze the development, mode of action, pharmacology, and structure-activity relationships of the currently known METTL3 inhibitors. They include both nucleoside and non-nucleoside compounds, with the latter comprising both competitive and allosteric inhibitors.- Published
- 2023
- Full Text
- View/download PDF
10. Potent and Specific Activators for Mitochondrial Sirtuins Sirt3 and Sirt5.
- Author
-
Suenkel B, Valente S, Zwergel C, Weiss S, Di Bello E, Fioravanti R, Aventaggiato M, Amorim JA, Garg N, Kumar S, Lombard DB, Hu T, Singh PK, Tafani M, Palmeira CM, Sinclair D, Mai A, and Steegborn C
- Subjects
- Humans, NAD, Sirtuin 1, Protein Isoforms metabolism, Peptides, Sirtuins metabolism, Sirtuin 3
- Abstract
Sirtuins are NAD
+ -dependent protein deacylases involved in metabolic regulation and aging-related diseases. Specific activators for seven human Sirtuin isoforms would be important chemical tools and potential therapeutic drugs. Activators have been described for Sirt1 and act via a unique N-terminal domain of this isoform. For most other Sirtuin isoforms, including mitochondrial Sirt3-5, no potent and specific activators have yet been identified. We here describe the identification and characterization of 1,4-dihydropyridine-based compounds that either act as pan Sirtuin activators or specifically stimulate Sirt3 or Sirt5. The activators bind to the Sirtuin catalytic cores independent of NAD+ and acylated peptides and stimulate turnover of peptide and protein substrates. The compounds also activate Sirt3 or Sirt5 in cellular systems regulating, e.g., apoptosis and electron transport chain. Our results provide a scaffold for potent Sirtuin activation and derivatives specific for Sirt3 and Sirt5 as an excellent basis for further drug development.- Published
- 2022
- Full Text
- View/download PDF
11. Polycomb Repressive Complex 2 Modulation through the Development of EZH2-EED Interaction Inhibitors and EED Binders.
- Author
-
Tomassi S, Romanelli A, Zwergel C, Valente S, and Mai A
- Subjects
- Animals, Antineoplastic Agents pharmacology, Cell Line, Tumor, Enhancer of Zeste Homolog 2 Protein metabolism, Enzyme Inhibitors pharmacology, Humans, Polycomb Repressive Complex 2 metabolism, Antineoplastic Agents therapeutic use, Enhancer of Zeste Homolog 2 Protein antagonists & inhibitors, Enzyme Inhibitors therapeutic use, Neoplasms drug therapy, Polycomb Repressive Complex 2 antagonists & inhibitors, Protein Binding drug effects
- Abstract
Epigenetics is nowadays a well-accepted area of research. In the last years, tremendous progress was made regarding molecules targeting EZH2, directly or indirectly. Recently tazemetostat hit the market after FDA-approval for the treatment of lymphoma. However, the impairment of EZH2 activity by orthosteric intervention has proven to be effective only in a limited subset of cancers. Considering the multiproteic nature of the PRC2 complex and the marked dependence of EZH2 functions on the other core subunits such as EED, in recent years, a new targeting approach ascended to prominence. The possibility to cripple the function of the PRC2 complex by interfering with its multimeric integrity fueled the interest in developing EZH2-EED protein-protein interaction and EED inhibitors as indirect modulators of PRC2-dependent methyltransferase activity. In this Perspective, we aim to summarize the latest findings regarding the development and the biological activity of these emerging classes of PRC2 modulators from a medicinal chemist's viewpoint.
- Published
- 2021
- Full Text
- View/download PDF
12. Structure-Guided Development of Small-Molecule PRC2 Inhibitors Targeting EZH2-EED Interaction.
- Author
-
Du D, Xu D, Zhu L, Stazi G, Zwergel C, Liu Y, Luo Z, Li Y, Zhang Y, Zhu K, Ding Y, Liu J, Fan S, Zhao K, Zhang N, Kong X, Jiang H, Chen K, Zhao K, Valente S, Min J, Duan W, and Luo C
- Subjects
- Cell Line, Tumor, Cell Proliferation drug effects, Drug Repositioning, Enhancer of Zeste Homolog 2 Protein metabolism, Enzyme Inhibitors chemical synthesis, Humans, Molecular Docking Simulation, Molecular Structure, Polycomb Repressive Complex 2 metabolism, Structure-Activity Relationship, Astemizole analogs & derivatives, Astemizole pharmacology, Enhancer of Zeste Homolog 2 Protein antagonists & inhibitors, Enzyme Inhibitors pharmacology, Polycomb Repressive Complex 2 antagonists & inhibitors, Protein Binding drug effects
- Abstract
Disruption of EZH2-embryonic ectoderm development (EED) protein-protein interaction (PPI) is a new promising cancer therapeutic strategy. We have previously reported the discovery of astemizole, a small-molecule inhibitor targeting the EZH2-EED PPI. Herein, we report the cocrystal structure of EED in complex with astemizole at 2.15 Å. The structure elucidates the detailed binding mode of astemizole to EED and provides a structure-guided design for the discovery of a novel EZH2-EED interaction inhibitor, DC-PRC2in-01, with an affinity K
d of 4.56 μM. DC-PRC2in-01 destabilizes the PRC2 complex, thereby leading to the degradation of PRC2 core proteins and the decrease of global H3K27me3 levels in cancer cells. The proliferation of PRC2-driven lymphomas cells is effectively inhibited, and the cell cycle is arrested in the G0/G1 phase. Together, these data demonstrate that DC-PRC2in-01 could be an effective chemical probe for investigating the PRC2-related physiology and pathology and providing a promising chemical scaffold for further development.- Published
- 2021
- Full Text
- View/download PDF
13. Discovery of the First Human Arylsulfatase A Reversible Inhibitor Impairing Mouse Oocyte Fertilization.
- Author
-
Caroselli S, Zwergel C, Pirolli A, Sabatino M, Xu Z, Kirsch G, Mai A, Colotti G, Altieri F, Canipari R, Valente S, and Ragno R
- Subjects
- Animals, Arylsulfatases metabolism, Cell Line, Tumor, Coumarins chemistry, Drug Discovery, Enzyme Inhibitors chemistry, Female, Humans, Male, Mice, Molecular Docking Simulation, Oocytes physiology, Sperm Motility drug effects, Spermatozoa drug effects, Spermatozoa physiology, Arylsulfatases antagonists & inhibitors, Coumarins pharmacology, Enzyme Inhibitors pharmacology, Fertilization drug effects, Oocytes drug effects
- Abstract
Arylsulfatase A (ARSA) plays a crucial role in the reproduction of mammals due to its involvement in the specific gamete interaction preceding sperm and egg fusion leading to fertilization. Recently, it has been shown that zona pellucida (ZP) sperm binding and in vivo fertilization in mice are markedly hampered by using a specific anti-ARSA antibody. Herein, the design and discovery of the first ARSA small molecule inhibitor based on a coumarin-containing polycycle are presented. Through a structure-based approach applied on our in-house library, compound 1r was identified as an ARSA reversible inhibitor (ARSAi); then its activity was validated through both surface plasmon resonance and biochemical inhibition experiments, the first providing a K
D value of 21 μM and the latter an IC50 value of 13.2 μM. Further investigations highlighted that compound 1r induced 20% sperm death at 25 μM and also impaired sperm motility; nevertheless both the effects were mediated by ROS production, since they were rescued by the cotreatment of 1r and N -acetyl cysteine (NAC). Interestingly, while 1r was not able to hamper the ZP/sperm binding, it markedly decreased the in vitro /NAC coadministration, hence allowing the ruling out of an ROS-dependent mechanism. In conclusion, herein is reported the first ever hit of ARSAi as a chemical tool that will enable better exploration of ARSA's biological role in fertilization as well as provide a starting point for developing 1r /NAC coadministration, hence allowing the ruling out of an ROS-dependent mechanism. In conclusion, herein is reported the first ever hit of ARSAi as a chemical tool that will enable better exploration of ARSA's biological role in fertilization as well as provide a starting point for developing 1r structure optimization aimed at increasing enzyme inhibition potency but also providing a deeper understanding of the involvement of ARSA in the fertilization pathway mechanism.- Published
- 2020
- Full Text
- View/download PDF
14. Design of First-in-Class Dual EZH2/HDAC Inhibitor: Biochemical Activity and Biological Evaluation in Cancer Cells.
- Author
-
Romanelli A, Stazi G, Fioravanti R, Zwergel C, Di Bello E, Pomella S, Perrone C, Battistelli C, Strippoli R, Tripodi M, Del Bufalo D, Rota R, Trisciuoglio D, Mai A, and Valente S
- Abstract
Since the histone modifying enzymes EZH2 and HDACs control a number of epigenetic-dependent carcinogenic pathways, we designed the first-in-class dual EZH2/HDAC inhibitor 5 displaying (sub)micromolar inhibition against both targets. When tested in several cancer cell lines, the hybrid 5 impaired cell viability at low micromolar level and in leukemia U937 and rhabdomyosarcoma RH4 cells provided G1 arrest, apoptotic induction, and increased differentiation, associated with an increase of acetyl-H3 and acetyl-α-tubulin and a decrease of H3K27me3 levels. In glioblastoma U87 cells, 5 hampered epithelial to mesenchymal transition by increasing the E-cadherin expression, thus proposing itself as a useful candidate for anticancer therapy., Competing Interests: The authors declare no competing financial interest., (Copyright © 2020 American Chemical Society.)
- Published
- 2020
- Full Text
- View/download PDF
15. Differential Network Analysis Reveals Metabolic Determinants Associated with Mortality in Acute Myocardial Infarction Patients and Suggests Potential Mechanisms Underlying Different Clinical Scores Used To Predict Death.
- Author
-
Vignoli A, Tenori L, Giusti B, Valente S, Carrabba N, Balzi D, Barchielli A, Marchionni N, Gensini GF, Marcucci R, Gori AM, Luchinat C, and Saccenti E
- Subjects
- Cohort Studies, Humans, Magnetic Resonance Spectroscopy, Metabolic Networks and Pathways, Metabolomics, Risk Factors, Myocardial Infarction
- Abstract
We present here the differential analysis of metabolite-metabolite association networks constructed from an array of 24 serum metabolites identified and quantified via nuclear magnetic resonance spectroscopy in a cohort of 825 patients of which 123 died within 2 years from acute myocardial infarction (AMI). We investigated differences in metabolite connectivity of patients who survived, at 2 years, the AMI event, and we characterized metabolite-metabolite association networks specific to high and low risks of death according to four different risk parameters, namely, acute coronary syndrome classification, Killip, Global Registry of Acute Coronary Events risk score, and metabolomics NOESY RF risk score. We show significant differences in the connectivity patterns of several low-molecular-weight molecules, implying variations in the regulation of several metabolic pathways regarding branched-chain amino acids, alanine, creatinine, mannose, ketone bodies, and energetic metabolism. Our results demonstrate that the characterization of metabolite-metabolite association networks is a promising and powerful tool to investigate AMI patients according to their outcomes at a molecular level.
- Published
- 2020
- Full Text
- View/download PDF
16. Discovery of a Novel Inhibitor of Histone Lysine-Specific Demethylase 1A (KDM1A/LSD1) as Orally Active Antitumor Agent.
- Author
-
Vianello P, Botrugno OA, Cappa A, Dal Zuffo R, Dessanti P, Mai A, Marrocco B, Mattevi A, Meroni G, Minucci S, Stazi G, Thaler F, Trifiró P, Valente S, Villa M, Varasi M, and Mercurio C
- Subjects
- Administration, Oral, Antineoplastic Agents administration & dosage, Antineoplastic Agents pharmacology, Cell Line, Tumor, Enzyme Inhibitors administration & dosage, Enzyme Inhibitors chemistry, Enzyme Inhibitors pharmacology, Enzyme Inhibitors therapeutic use, Histone Demethylases metabolism, Humans, Leukemia, Promyelocytic, Acute metabolism, Structure-Activity Relationship, Tranylcypromine administration & dosage, Tranylcypromine pharmacology, Antineoplastic Agents chemistry, Antineoplastic Agents therapeutic use, Histone Demethylases antagonists & inhibitors, Leukemia, Promyelocytic, Acute drug therapy, Tranylcypromine chemistry, Tranylcypromine therapeutic use
- Abstract
We report the stereoselective synthesis and biological activity of a novel series of tranylcypromine (TCPA) derivatives (14a-k, 15, 16), potent inhibitors of KDM1A. The new compounds strongly inhibit the clonogenic potential of acute leukemia cell lines. In particular three molecules (14d, 14e, and 14g) showing selectivity versus MAO A and remarkably inhibiting colony formation in THP-1 human leukemia cells, were assessed in mouse for their preliminary pharmacokinetic. 14d and 14e were further tested in vivo in a murine acute promyelocytic leukemia model, resulting 14d the most effective. Its two enantiomers were synthesized: the (1S,2R) enantiomer 15 showed higher activity than its (1R,2S) analogue 16, in both biochemical and cellular assays. Compound 15 exhibited in vivo efficacy after oral administration, determining a 62% increased survival in mouse leukemia model with evidence of KDM1A inhibition. The biological profile of compound 15 supports its further investigation as a cancer therapeutic.
- Published
- 2016
- Full Text
- View/download PDF
17. 1,4-Dihydropyridines Active on the SIRT1/AMPK Pathway Ameliorate Skin Repair and Mitochondrial Function and Exhibit Inhibition of Proliferation in Cancer Cells.
- Author
-
Valente S, Mellini P, Spallotta F, Carafa V, Nebbioso A, Polletta L, Carnevale I, Saladini S, Trisciuoglio D, Gabellini C, Tardugno M, Zwergel C, Cencioni C, Atlante S, Moniot S, Steegborn C, Budriesi R, Tafani M, Del Bufalo D, Altucci L, Gaetano C, and Mai A
- Subjects
- Animals, Antineoplastic Agents chemistry, Cell Line, Cell Line, Tumor, Cell Proliferation drug effects, Dihydropyridines chemistry, Enzyme Activation drug effects, Humans, Male, Mice, Mitochondria metabolism, Mitochondria pathology, Neoplasms drug therapy, Neoplasms metabolism, Neoplasms pathology, Signal Transduction drug effects, Skin metabolism, Skin pathology, AMP-Activated Protein Kinases metabolism, Antineoplastic Agents pharmacology, Dihydropyridines pharmacology, Mitochondria drug effects, Sirtuin 1 metabolism, Skin drug effects, Wound Healing drug effects
- Abstract
Modulators of sirtuins are considered promising therapeutic targets for the treatment of cancer, cardiovascular, metabolic, inflammatory, and neurodegenerative diseases. Here we prepared new 1,4-dihydropyridines (DHPs) bearing changes at the C2/C6, C3/C5, C4, or N1 position. Tested with the SIRTainty procedure, some of them displayed increased SIRT1 activation with respect to the prototype 3a, high NO release in HaCat cells, and ameliorated skin repair in a mouse model of wound healing. In C2C12 myoblasts, two of them improved mitochondrial density and functions. All the effects were reverted by coadministration of compound C (9), an AMPK inhibitor, or of EX-527 (10), a SIRT1 inhibitor, highlighting the involvement of the SIRT1/AMPK pathway in the action of DHPs. Finally, tested in a panel of cancer cells, the water-soluble form of 3a, compound 8, displayed antiproliferative effects in the range of 8-35 μM and increased H4K16 deacetylation, suggesting a possible role for SIRT1 activators in cancer therapy.
- Published
- 2016
- Full Text
- View/download PDF
18. Discovery of Inhibitors for the Ether Lipid-Generating Enzyme AGPS as Anti-Cancer Agents.
- Author
-
Piano V, Benjamin DI, Valente S, Nenci S, Marrocco B, Mai A, Aliverti A, Nomura DK, and Mattevi A
- Subjects
- Animals, Antineoplastic Agents chemistry, Antineoplastic Agents pharmacology, Crystallography, X-Ray, Drug Stability, Enzyme Inhibitors chemistry, Enzyme Inhibitors pharmacology, Humans, Models, Molecular, Molecular Structure, Swine, Temperature, Alkyl and Aryl Transferases antagonists & inhibitors, Drug Discovery, Small Molecule Libraries chemistry, Small Molecule Libraries pharmacology
- Abstract
Dysregulated ether lipid metabolism is an important hallmark of cancer cells. Previous studies have reported that lowering ether lipid levels by genetic ablation of the ether lipid-generating enzyme alkyl-glycerone phosphate synthase (AGPS) lowers key structural and oncogenic ether lipid levels and alters fatty acid, glycerophospholipid, and eicosanoid metabolism to impair cancer pathogenicity, indicating that AGPS may be a potential therapeutic target for cancer. In this study, we have performed a small-molecule screen to identify candidate AGPS inhibitors. We have identified several lead AGPS inhibitors and have structurally characterized their interactions with the enzyme and show that these inhibitors bind to distinct portions of the active site. We further show that the lead AGPS inhibitor 1a selectively lowers ether lipid levels in several types of human cancer cells and impairs their cellular survival and migration. We provide here the first report of in situ-active pharmacological tools for inhibiting AGPS, which may provide chemical scaffolds for future AGPS inhibitor development for cancer therapy.
- Published
- 2015
- Full Text
- View/download PDF
19. Novel histone deacetylase inhibitors induce growth arrest, apoptosis, and differentiation in sarcoma cancer stem cells.
- Author
-
Di Pompo G, Salerno M, Rotili D, Valente S, Zwergel C, Avnet S, Lattanzi G, Baldini N, and Mai A
- Subjects
- Aminopyridines chemical synthesis, Aminopyridines pharmacology, Bone Neoplasms pathology, Cell Line, Tumor, Drug Screening Assays, Antitumor, Histone Deacetylase Inhibitors chemical synthesis, Histone Deacetylase Inhibitors pharmacology, Humans, Hydroxamic Acids chemical synthesis, Hydroxamic Acids pharmacology, Neoplastic Stem Cells pathology, Osteogenesis, Osteosarcoma pathology, Pyrimidinones chemical synthesis, Pyrimidinones pharmacology, Rhabdomyosarcoma pathology, Sarcoma drug therapy, Sarcoma, Ewing pathology, Structure-Activity Relationship, Aminopyridines chemistry, Apoptosis drug effects, Cell Differentiation drug effects, Cell Proliferation drug effects, Histone Deacetylase Inhibitors chemistry, Hydroxamic Acids chemistry, Neoplastic Stem Cells drug effects, Pyrimidinones chemistry, Sarcoma pathology
- Abstract
Musculoskeletal sarcomas are aggressive malignancies of bone and soft tissues often affecting children and adolescents. Histone deacetylase inhibitors (HDACi) have been proposed to counteract cancer stem cells (CSCs) in solid neoplasms. When tested in human osteosarcoma, rhabdomyosarcoma, and Ewing's sarcoma stem cells, the new HDACi MC1742 (1) and MC2625 (2) increased acetyl-H3 and acetyl-tubulin levels and inhibited CSC growth by apoptosis induction. At nontoxic doses, 1 promoted osteogenic differentiation. Further investigation with 1 will be done in preclinical sarcoma models.
- Published
- 2015
- Full Text
- View/download PDF
20. Pure Diastereomers of a Tranylcypromine-Based LSD1 Inhibitor: Enzyme Selectivity and In-Cell Studies.
- Author
-
Valente S, Rodriguez V, Mercurio C, Vianello P, Saponara B, Cirilli R, Ciossani G, Labella D, Marrocco B, Ruoppolo G, Botrugno OA, Dessanti P, Minucci S, Mattevi A, Varasi M, and Mai A
- Abstract
The pure four diastereomers (11a-d) of trans-benzyl (1-((4-(2-aminocyclopropyl)phenyl)amino)-1-oxo-3-phenylpropan-2-yl)carbamate hydrochloride 11, previously described by us as LSD1 inhibitor, were obtained by enantiospecific synthesis/chiral HPLC separation method. Tested in LSD1 and MAO assays, 11b (S,1S,2R) and 11d (R,1S,2R) were the most potent isomers against LSD1 and were less active against MAO-A and practically inactive against MAO-B. In cells, all the four diastereomers induced Gfi-1b and ITGAM gene expression in NB4 cells, accordingly with their LSD1 inhibition, and 11b and 11d inhibited the colony forming potential in murine promyelocytic blasts.
- Published
- 2014
- Full Text
- View/download PDF
21. 1,3,4-Oxadiazole-containing histone deacetylase inhibitors: anticancer activities in cancer cells.
- Author
-
Valente S, Trisciuoglio D, De Luca T, Nebbioso A, Labella D, Lenoci A, Bigogno C, Dondio G, Miceli M, Brosch G, Del Bufalo D, Altucci L, and Mai A
- Subjects
- Antineoplastic Agents chemical synthesis, Antineoplastic Agents chemistry, Cell Proliferation drug effects, Dose-Response Relationship, Drug, Doxorubicin pharmacology, Drug Screening Assays, Antitumor, HCT116 Cells, HL-60 Cells, Histone Deacetylase Inhibitors chemical synthesis, Histone Deacetylase Inhibitors chemistry, Humans, Molecular Structure, Oxadiazoles chemical synthesis, Oxadiazoles chemistry, Structure-Activity Relationship, Tumor Cells, Cultured, Antineoplastic Agents pharmacology, Histone Deacetylase Inhibitors pharmacology, Oxadiazoles pharmacology
- Abstract
We describe 1,3,4-oxadiazole-containing hydroxamates (2) and 2-aminoanilides (3) as histone deacetylase inhibitors. Among them, 2t, 2x, and 3i were the most potent and selective against HDAC1. In U937 leukemia cells, 2t was more potent than SAHA in inducing apoptosis, and 3i displayed cell differentiation with a potency similar to MS-275. In several acute myeloid leukemia (AML) cell lines, as well as in U937 cells in combination with doxorubicin, 3i showed higher antiproliferative effects than SAHA.
- Published
- 2014
- Full Text
- View/download PDF
22. Selective non-nucleoside inhibitors of human DNA methyltransferases active in cancer including in cancer stem cells.
- Author
-
Valente S, Liu Y, Schnekenburger M, Zwergel C, Cosconati S, Gros C, Tardugno M, Labella D, Florean C, Minden S, Hashimoto H, Chang Y, Zhang X, Kirsch G, Novellino E, Arimondo PB, Miele E, Ferretti E, Gulino A, Diederich M, Cheng X, and Mai A
- Subjects
- Aminoquinolines chemistry, Aminoquinolines pharmacology, Animals, Antineoplastic Agents chemistry, Antineoplastic Agents pharmacology, Benzamides chemistry, Benzamides pharmacology, Cell Line, Tumor, Drug Screening Assays, Antitumor, Humans, Mice, Pyrimidines chemistry, Pyrimidines pharmacology, Quinolines chemistry, Quinolines pharmacology, Structure-Activity Relationship, Aminoquinolines chemical synthesis, Antineoplastic Agents chemical synthesis, Benzamides chemical synthesis, DNA (Cytosine-5-)-Methyltransferases antagonists & inhibitors, Neoplastic Stem Cells drug effects, Pyrimidines chemical synthesis, Quinolines chemical synthesis
- Abstract
DNA methyltransferases (DNMTs) are important enzymes involved in epigenetic control of gene expression and represent valuable targets in cancer chemotherapy. A number of nucleoside DNMT inhibitors (DNMTi) have been studied in cancer, including in cancer stem cells, and two of them (azacytidine and decitabine) have been approved for treatment of myelodysplastic syndromes. However, only a few non-nucleoside DNMTi have been identified so far, and even fewer have been validated in cancer. Through a process of hit-to-lead optimization, we report here the discovery of compound 5 as a potent non-nucleoside DNMTi that is also selective toward other AdoMet-dependent protein methyltransferases. Compound 5 was potent at single-digit micromolar concentrations against a panel of cancer cells and was less toxic in peripheral blood mononuclear cells than two other compounds tested. In mouse medulloblastoma stem cells, 5 inhibited cell growth, whereas related compound 2 showed high cell differentiation. To the best of our knowledge, 2 and 5 are the first non-nucleoside DNMTi tested in a cancer stem cell line.
- Published
- 2014
- Full Text
- View/download PDF
23. Pan-histone demethylase inhibitors simultaneously targeting Jumonji C and lysine-specific demethylases display high anticancer activities.
- Author
-
Rotili D, Tomassi S, Conte M, Benedetti R, Tortorici M, Ciossani G, Valente S, Marrocco B, Labella D, Novellino E, Mattevi A, Altucci L, Tumber A, Yapp C, King ON, Hopkinson RJ, Kawamura A, Schofield CJ, and Mai A
- Subjects
- Antineoplastic Agents pharmacology, Apoptosis drug effects, Cell Line, Tumor, Enzyme Inhibitors pharmacology, Humans, Molecular Docking Simulation, Structure-Activity Relationship, Antineoplastic Agents chemical synthesis, Enzyme Inhibitors chemical synthesis, Histone Demethylases antagonists & inhibitors, Jumonji Domain-Containing Histone Demethylases antagonists & inhibitors
- Abstract
In prostate cancer, two different types of histone lysine demethylases (KDM), LSD1/KDM1 and JMJD2/KDM4, are coexpressed and colocalize with the androgen receptor. We designed and synthesized hybrid LSD1/JmjC or "pan-KDM" inhibitors 1-6 by coupling the skeleton of tranylcypromine 7, a known LSD1 inhibitor, with 4-carboxy-4'-carbomethoxy-2,2'-bipyridine 8 or 5-carboxy-8-hydroxyquinoline 9, two 2-oxoglutarate competitive templates developed for JmjC inhibition. Hybrid compounds 1-6 are able to simultaneously target both KDM families and have been validated as potential antitumor agents in cells. Among them, 2 and 3 increase H3K4 and H3K9 methylation levels in cells and cause growth arrest and substantial apoptosis in LNCaP prostate and HCT116 colon cancer cells. When tested in noncancer mesenchymal progenitor (MePR) cells, 2 and 3 induced little and no apoptosis, respectively, thus showing cancer-selective inhibiting action.
- Published
- 2014
- Full Text
- View/download PDF
24. Novel reversible monoamine oxidase A inhibitors: highly potent and selective 3-(1H-pyrrol-3-yl)-2-oxazolidinones.
- Author
-
Valente S, Tomassi S, Tempera G, Saccoccio S, Agostinelli E, and Mai A
- Subjects
- Animals, Cattle, In Vitro Techniques, Isoenzymes antagonists & inhibitors, Isoenzymes metabolism, Monoamine Oxidase Inhibitors chemistry, Monoamine Oxidase Inhibitors pharmacology, Oxazolidinones chemistry, Oxazolidinones pharmacology, Pyrroles chemistry, Pyrroles pharmacology, Stereoisomerism, Structure-Activity Relationship, Monoamine Oxidase metabolism, Monoamine Oxidase Inhibitors chemical synthesis, Oxazolidinones chemical synthesis, Pyrroles chemical synthesis
- Abstract
Monoamine oxidases (MAOs) are involved in various psychiatric and neurodegenerative disorders; hence, MAO inhibitors are useful agents in the therapy of Parkinson's disease, Alzheimer's dementia, and depression syndrome. Herein we report a novel series of 3-(1H-pyrrol-3-yl)-2-oxazolidinones 3-7 as reversible, highly potent and selective anti-MAO-A agents. In particular, 4b, 5b, and 4c showed a K(i-MAO-A) of 0.6, 0.8, and 1 nM, respectively, 4c being 200000-fold selective for MAO-A with respect to MAO-B.
- Published
- 2011
- Full Text
- View/download PDF
25. Novel 3,5-bis(bromohydroxybenzylidene)piperidin-4-ones as coactivator-associated arginine methyltransferase 1 inhibitors: enzyme selectivity and cellular activity.
- Author
-
Cheng D, Valente S, Castellano S, Sbardella G, Di Santo R, Costi R, Bedford MT, and Mai A
- Subjects
- Cell Line, Tumor, Cell Survival drug effects, Epigenesis, Genetic, HEK293 Cells, Humans, Luciferases genetics, Luciferases metabolism, Methylation, Piperidines chemistry, Piperidines pharmacology, Poly(A)-Binding Protein I genetics, Poly(A)-Binding Protein I metabolism, Promoter Regions, Genetic, Prostate-Specific Antigen genetics, Protein-Arginine N-Methyltransferases genetics, Structure-Activity Relationship, Piperidines chemical synthesis, Protein-Arginine N-Methyltransferases antagonists & inhibitors
- Abstract
Coactivator-associated arginine methyltransferase 1 (CARM1) represents a valuable target for hormone-dependent tumors such as prostate and breast cancers. Here we report the enzyme and cellular characterization of the 1-benzyl-3,5-bis(3-bromo-4-hydroxybenzylidene)piperidin-4-one (7g) and its analogues 8a-l. Among them, 7g, 8e, and 8l displayed high and selective CARM1 inhibition, with lower or no activity against a panel of different PRMTs or HKMTs. In human LNCaP cells, 7g showed a significant dose-dependent reduction of the PSA promoter activity.
- Published
- 2011
- Full Text
- View/download PDF
26. Biochemical, structural, and biological evaluation of tranylcypromine derivatives as inhibitors of histone demethylases LSD1 and LSD2.
- Author
-
Binda C, Valente S, Romanenghi M, Pilotto S, Cirilli R, Karytinos A, Ciossani G, Botrugno OA, Forneris F, Tardugno M, Edmondson DE, Minucci S, Mattevi A, and Mai A
- Subjects
- Animals, Antineoplastic Agents pharmacology, Cell Line, Drug Synergism, Histone Demethylases chemistry, Humans, Mice, Models, Molecular, Molecular Conformation, Stereoisomerism, Substrate Specificity, Enzyme Inhibitors chemistry, Enzyme Inhibitors pharmacology, Histone Demethylases antagonists & inhibitors, Tranylcypromine chemistry, Tranylcypromine pharmacology
- Abstract
LSD1 and LSD2 histone demethylases are implicated in a number of physiological and pathological processes, ranging from tumorigenesis to herpes virus infection. A comprehensive structural, biochemical, and cellular study is presented here to probe the potential of these enzymes for epigenetic therapies. This approach employs tranylcypromine as a chemical scaffold for the design of novel demethylase inhibitors. This drug is a clinically validated antidepressant known to target monoamine oxidases A and B. These two flavoenzymes are structurally related to LSD1 and LSD2. Mechanistic and crystallographic studies of tranylcypromine inhibition reveal a lack of selectivity and differing covalent modifications of the FAD cofactor depending on the enantiomeric form. These findings are pharmacologically relevant, since tranylcypromine is currently administered as a racemic mixture. A large set of tranylcypromine analogues were synthesized and screened for inhibitory activities. We found that the common evolutionary origin of LSD and MAO enzymes, despite their unrelated functions and substrate specificities, is reflected in related ligand-binding properties. A few compounds with partial enzyme selectivity were identified. The biological activity of one of these new inhibitors was evaluated with a cellular model of acute promyelocytic leukemia chosen since its pathogenesis includes aberrant activities of several chromatin modifiers. Marked effects on cell differentiation and an unprecedented synergistic activity with antileukemia drugs were observed. These data demonstrate that these LSD1/2 inhibitors are of potential relevance for the treatment of promyelocytic leukemia and, more generally, as tools to alter chromatin state with promise of a block of tumor progression.
- Published
- 2010
- Full Text
- View/download PDF
27. Study of 1,4-dihydropyridine structural scaffold: discovery of novel sirtuin activators and inhibitors.
- Author
-
Mai A, Valente S, Meade S, Carafa V, Tardugno M, Nebbioso A, Galmozzi A, Mitro N, De Fabiani E, Altucci L, and Kazantsev A
- Subjects
- Alkynes chemistry, Amines chemistry, Animals, Benzaldehydes chemistry, Cell Line, Tumor, Enzyme Activation drug effects, Enzyme Inhibitors chemical synthesis, Humans, Mesenchymal Stem Cells cytology, Mesenchymal Stem Cells enzymology, Mice, Myoblasts cytology, Myoblasts enzymology, Propionates chemistry, Pyridines chemical synthesis, Enzyme Inhibitors chemistry, Enzyme Inhibitors pharmacology, Pyridines chemistry, Pyridines pharmacology, Sirtuins antagonists & inhibitors, Sirtuins metabolism
- Abstract
NAD(+)-dependent sirtuin deacetylases have emerged as potential therapeutic targets for treatment of human illnesses such as cancer, metabolic, cardiovascular, and neurodegenerative diseases. The benefits of sirtuin modulation by small molecules have been demonstrated for these diseases. In contrast to the discovery of inhibitors of SIRT1, -2, and -3, only activators for SIRT1 are known. Here, we rationalized the potential of the previously unexplored dihydropyridine scaffold in developing sirtuin ligands, thus we prepared a series of 1,4-dihydropyridine-based derivatives 1-3. Assessment of their SIRT1-3 deacetylase activities revealed the importance of the substituent at the N1 position of the dihydropyridine structure on sirtuin activity. Placement of cyclopropyl, phenyl, or phenylethyl groups at N1 conferred nonselective SIRT1 and SIRT2 inhibition activity, while a benzyl group at N1 conferred potent SIRT1, -2, and -3 activation. Senescence assays performed on hMSC and mitochondrial function studies conducted with murine C2C12 myoblasts confirmed the compounds' novel and unique SIRT-activating properties.
- Published
- 2009
- Full Text
- View/download PDF
28. epigenetic multiple ligands: mixed histone/protein methyltransferase, acetyltransferase, and class III deacetylase (sirtuin) inhibitors.
- Author
-
Mai A, Cheng D, Bedford MT, Valente S, Nebbioso A, Perrone A, Brosch G, Sbardella G, De Bellis F, Miceli M, and Altucci L
- Subjects
- Apoptosis drug effects, Cell Cycle drug effects, Cell Differentiation drug effects, Cell Line, Tumor, Drug Evaluation, Preclinical, Drug Screening Assays, Antitumor, Eosine Yellowish-(YS) chemistry, Granulocytes drug effects, Histone Deacetylases, Humans, Ligands, Molecular Structure, Sirtuins antagonists & inhibitors, Stereoisomerism, Structure-Activity Relationship, Acetyltransferases antagonists & inhibitors, Eosine Yellowish-(YS) analogs & derivatives, Eosine Yellowish-(YS) pharmacology, Histone Deacetylase Inhibitors, Histones antagonists & inhibitors, Methyltransferases antagonists & inhibitors
- Abstract
A number of new compounds bearing two ortho-bromo- and ortho, ortho-dibromophenol moieties linked through a saturated/unsaturated, linear/(poly)cyclic spacer (compounds 1- 9) were prepared as simplified analogues of AMI-5 (eosin), a recently reported inhibitor of both protein arginine and histone lysine methyltransferases (PRMTs and HKMTs). Such compounds were tested against a panel of PRMTs (RmtA, PRMT1, and CARM1) and against human SET7 (a HKMT), using histone and nonhistone proteins as a substrate. They were also screened against HAT and SIRTs, because they are structurally related to some HAT and/or SIRT modulators. From the inhibitory data, some of tested compounds ( 1b, 1c, 4b, 4f, 4j, 4l, 7b, and 7f) were able to inhibit PRMTs, HKMT, HAT, and SIRTs with similar potency, thus behaving as multiple ligands for these epigenetic targets (epi-MLs). When tested on the human leukemia U937 cell line, the epi-MLs induced high apoptosis levels [i.e., 40.7% ( 4l) and 42.6% ( 7b)] and/or massive, dose-dependent cytodifferentiation [i.e., 95.2% ( 1c) and 96.1% ( 4j)], whereas the single-target inhibitors eosin, curcumin, and sirtinol were ineffective or showed a weak effect.
- Published
- 2008
- Full Text
- View/download PDF
29. 3-D QSAR studies on histone deacetylase inhibitors. A GOLPE/GRID approach on different series of compounds.
- Author
-
Ragno R, Simeoni S, Valente S, Massa S, and Mai A
- Subjects
- Models, Molecular, Quantitative Structure-Activity Relationship, Enzyme Inhibitors chemistry, Enzyme Inhibitors pharmacology, Histone Deacetylase Inhibitors
- Abstract
Docking simulation and three-dimensional quantitative structure-activity relationships (3D-QSARs) analyses were conducted on four series of HDAC inhibitors. The studies were performed using the GRID/GOLPE combination using structure-based alignment. Twelve 3-D QSAR models were derived and discussed. Compared to previous studies on similar inhibitors, the present 3-D QSAR investigation proved to be of higher statistical value, displaying for the best global model r2, q2, and cross-validated SDEP values of 0.94, 0.83, and 0.41, respectively. A comparison of the 3-D QSAR maps with the structural features of the binding site showed good correlation. The results of 3D-QSAR and docking studies validated each other and provided insight into the structural requirements for anti-HDAC activity. To our knowledge this is the first 3-D QSAR application on a broad molecular diversity training set of HDACIs.
- Published
- 2006
- Full Text
- View/download PDF
30. 3-(4-Aroyl-1-methyl-1H-pyrrol-2-yl)-N-hydroxy-2-propenamides as a new class of synthetic histone deacetylase inhibitors. 3. Discovery of novel lead compounds through structure-based drug design and docking studies.
- Author
-
Ragno R, Mai A, Massa S, Cerbara I, Valente S, Bottoni P, Scatena R, Jesacher F, Loidl P, and Brosch G
- Subjects
- Amides chemistry, Amides pharmacology, Animals, Antineoplastic Agents chemistry, Antineoplastic Agents pharmacology, Catalytic Domain, Cell Differentiation drug effects, Cell Division drug effects, Cell Line, Tumor, Drug Design, Hemoglobins metabolism, Histone Deacetylases chemistry, Mice, Models, Molecular, Propionates chemistry, Propionates pharmacology, Protein Binding, Pyrroles chemistry, Pyrroles pharmacology, Structure-Activity Relationship, Zea mays, Amides chemical synthesis, Antineoplastic Agents chemical synthesis, Histone Deacetylase Inhibitors, Propionates chemical synthesis, Pyrroles chemical synthesis
- Abstract
Aroyl-pyrrole-hydroxy-amides (APHAs) are a new class of synthetic HDAC inhibitors recently described by us. Through three different docking procedures we designed, synthesized, and tested two new isomers of APHA lead compound 3-(4-benzoyl-1-methyl-1H-pyrrol-2-yl)-N-hydroxy-2-propenamide (1), compounds 3 and 4, characterized by different insertions of benzoyl and propenoylhydroxamate groups onto the pyrrole ring. Biological activities of 3 and 4 were predicted by computational tools up to 617-fold more potent than that of 1 against HDAC1; thus, 3 and 4 were synthesized and tested against both mouse HDAC1 and maize HD2 enzymes. Predictions of biological affinities (K(i) values) of 3 and 4, performed by a VALIDATE model (applied on either SAD or automatic DOCK or Autodock results) and by the Autodock internal scoring function, were in good agreement with experimental activities. Ligand/receptor positive interactions made by 3 and 4 into the catalytic pocket, in addition to those showed by 1, could at least in part account for their higher HDAC1 inhibitory activities. In particular, in mouse HDAC1 inhibitory assay 3 and 4 were 19- and 6-times more potent than 1, respectively, and 3 and 4 antimaize HD2 activities were 16- and 76-times higher than that of 1, 4 being as potent as SAHA in this assay. Compound 4, tested as antiproliferative and cytodifferentiating agent on MEL cells, showed dose-dependent growth inhibition and hemoglobin accumulation effects.
- Published
- 2004
- Full Text
- View/download PDF
31. 3-(4-Aroyl-1-methyl-1H-2-pyrrolyl)-N-hydroxy-2-propenamides as a new class of synthetic histone deacetylase inhibitors. 2. Effect of pyrrole-C2 and/or -C4 substitutions on biological activity.
- Author
-
Mai A, Massa S, Cerbara I, Valente S, Ragno R, Bottoni P, Scatena R, Loidl P, and Brosch G
- Subjects
- Acrylamides chemistry, Acrylamides pharmacology, Animals, Antineoplastic Agents chemistry, Antineoplastic Agents pharmacology, Cell Differentiation drug effects, Cell Division drug effects, Cell Line, Tumor, Drug Screening Assays, Antitumor, Mice, Models, Molecular, Molecular Conformation, Pyrroles chemistry, Pyrroles pharmacology, Structure-Activity Relationship, Acrylamides chemical synthesis, Antineoplastic Agents chemical synthesis, Histone Deacetylase Inhibitors, Pyrroles chemical synthesis
- Abstract
Previous SAR studies (Part 1: Mai, A.; et al. J. Med. Chem. 2003, 46, 512-524) performed on some portions (pyrrole-C4, pyrrole-N1, and hydroxamate group) of 3-(4-benzoyl-1-methyl-1H-pyrrol-2-yl)-N-hydroxy-2-propenamide (1a) highlighted its 4-phenylacetyl (1b) and 4-cynnamoyl (1c) analogues as more potent compounds in inhibiting maize HD2 activity in vitro. In the present paper, we investigated the effect on anti-HD2 activity of chemical substitutions performed on the pyrrole-C2 ethene chains of 1a-c, which were replaced with methylene, ethylene, substituted ethene, and 1,3-butadiene chains (compounds 2). Biological results clearly indicated the unsubstituted ethene chain as the best structural motif to get the highest HDAC inhibitory activity, the sole exception to this rule being the introduction of the 1,3-butadienyl moiety into the 1a chemical structure (IC50(2f) = 0.77 microM; IC50(1a) = 3.8 microM). IC50 values of compounds 3, prepared as 1b homologues, revealed that between benzene and carbonyl groups at the pyrrole-C(4) position a hydrocarbon spacer length ranging from two to five methylenes is well accepted by the APHA template, being that 3a (two methylenes) and 3d (five methylenes) are more potent (2.3- and 1.4-fold, respectively) than 1b, while the introduction of a higher number of methylene units (see 3e,f) decreased the inhibitory activities of the derivatives. Particularly, 3a (IC50 = 0.043 microM) showed the same potency as SAHA in inhibiting HD2 in vitro, and it was 3000- and 2.6-fold more potent than sodium valproate and HC-toxin and was 4.3- and 6-fold less potent than trapoxin and TSA, respectively. Finally, conformationally constrained forms of 1b,c (compounds 4), prepared with the aim to obtain some information potentially useful for a future 3D-QSAR study, showed the same (4a,b) or higher (4c,d) HD2 inhibiting activities in comparison with those of the reference drugs. Molecular modeling and docking calculations on the designed compounds performed in parallel with the chemistry work fully supported the synthetic effort and gave insights into the binding mode of the more flexible APHA derivatives (i.e., 3a). Despite the difference of potency between 1b and 3a in the enzyme assay, the two APHA derivatives showed similar antiproliferative and cytodifferentiating activities in vivo on Friends MEL cells, being that 3a is more potent than 1b in the differentiation assay only at the highest tested dose (48 microM).
- Published
- 2004
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.