Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Chem Inf Model ; 63(7): 2158-2169, 2023 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-36930801

RESUMEN

The rapid global spread of the SARS-CoV-2 virus facilitated the development of novel direct-acting antiviral agents (DAAs). The papain-like protease (PLpro) has been proposed as one of the major SARS-CoV-2 targets for DAAs due to its dual role in processing viral proteins and facilitating the host's immune suppression. This dual role makes identifying small molecules that can effectively neutralize SARS-CoV-2 PLpro activity a high-priority task. However, PLpro drug discovery faces a significant challenge due to the high mobility and induced-fit effects in the protease's active site. Herein, we virtually screened the ZINC20 database with Deep Docking (DD) to identify prospective noncovalent PLpro binders and combined ultra-large consensus docking with two pharmacophore (ph4)-filtering strategies. The analysis of active compounds revealed their somewhat-limited diversity, likely attributed to the induced-fit nature of PLpro's active site in the crystal structures, and therefore, the use of rigid docking protocols poses inherited limitations. The top hits were assessed against recombinant viral proteins and live viruses, demonstrating desirable inhibitory activities. The best compound VPC-300195 (IC50: 15 µM) ranks among the top noncovalent PLpro inhibitors discovered through in silico methodologies. In the search for novel SARS-CoV-2 PLpro-specific chemotypes, the identified inhibitors could serve as diverse templates for the development of effective noncovalent PLpro inhibitors.


Asunto(s)
COVID-19 , Hepatitis C Crónica , Humanos , SARS-CoV-2 , Antivirales/farmacología , Antivirales/química , Modelos Moleculares , Estudios Prospectivos , Inhibidores de Proteasas/farmacología , Inhibidores de Proteasas/química , Proteínas Virales/química , Péptido Hidrolasas
2.
Mol Ther ; 30(9): 3034-3051, 2022 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-35733339

RESUMEN

Approved drugs for the treatment of osteoporosis can prevent further bone loss but do not stimulate bone formation. Approaches that improve bone density in metabolic diseases are needed. Therapies that take advantage of the ability of mesenchymal stem cells (MSCs) to differentiate into various osteogenic lineages to treat bone disorders are of particular interest. Here we examine the ability of small interfering RNA (siRNA) to enhance osteoblast differentiation and bone formation by silencing the negative suppressor gene GNAS in bone MSCs. Using clinically validated lipid nanoparticle (LNP) siRNA delivery systems, we show that silencing the suppressor gene GNAS in vitro in MSCs leads to molecular and phenotypic changes similar to those seen in osteoblasts. Further, we demonstrate that these LNP-siRNAs can transfect a large proportion of mice MSCs in the compact bone following intravenous injection. Transfection of MSCs in various animal models led to silencing of GNAS and enhanced differentiation of MSCs into osteoblasts. These data demonstrate the potential for LNP delivery of siRNA to enhance the differentiation of MSCs into osteoblasts, and suggests that they are a promising approach for the treatment of osteoporosis and other bone diseases.


Asunto(s)
Células Madre Mesenquimatosas , Osteoporosis , Animales , Diferenciación Celular/genética , Células Cultivadas , Liposomas , Células Madre Mesenquimatosas/metabolismo , Ratones , Nanopartículas , Osteoblastos/metabolismo , Osteogénesis/genética , Osteoporosis/genética , Osteoporosis/metabolismo , Osteoporosis/terapia , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo
3.
Mol Pharmacol ; 94(2): 917-925, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29903751

RESUMEN

Deletion of phenylalanine at position 508 (F508del) in cystic fibrosis transmembrane conductance regulator (CFTR) is the most common cystic fibrosis (CF)-causing mutation. Recently, ORKAMBI, a combination therapy that includes a corrector of the processing defect of F508del-CFTR (lumacaftor or VX-809) and a potentiator of channel activity (ivacaftor or VX-770), was approved for CF patients homozygous for this mutation. However, clinical studies revealed that the effect of ORKAMBI on lung function is modest and it was proposed that this modest effect relates to a negative impact of VX-770 on the stability of F508del-CFTR. In the current studies, we showed that this negative effect of VX-770 at 10 µM correlated with its inhibitory effect on VX-809-mediated correction of the interface between the second membrane spanning domain and the first nucleotide binding domain bearing F508del. Interestingly, we found that VX-770 exerted a similar negative effect on the stability of other membrane localized solute carriers (SLC26A3, SLC26A9, and SLC6A14), suggesting that this negative effect is not specific for F508del-CFTR. We determined that the relative destabilizing effect of a panel of VX-770 derivatives on F508del-CFTR correlated with their predicted lipophilicity. Polarized total internal reflection fluorescence microscopy on a supported lipid bilayer model shows that VX-770, and not its less lipophilic derivative, increased the fluidity of and reorganized the membrane. In summary, our findings show that there is a potential for nonspecific effects of VX-770 on the lipid bilayer and suggest that this effect may account for its destabilizing effect on VX-809- rescued F508del-CFTR.


Asunto(s)
Aminofenoles/farmacología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/química , Fibrosis Quística/genética , Quinolonas/farmacología , Transportadores de Sulfato/química , Aminofenoles/química , Aminopiridinas/farmacología , Benzodioxoles/farmacología , Línea Celular , Fibrosis Quística/tratamiento farmacológico , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Liofilización , Células HEK293 , Humanos , Microscopía Fluorescente , Mutación , Estabilidad Proteica/efectos de los fármacos , Quinolonas/química
4.
Bioconjug Chem ; 26(6): 1095-103, 2015 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-25945831

RESUMEN

The in vivo hydrolytic pathway of a dual-function bone-targeting EP4 receptor agonist-bisphosphonate pro-drug was deduced from radiolabeling experiments. A (14)C labeled pro-drug was used to monitor liberation of the bisphosphonate and results were compared to parallel studies where the EP4 receptor agonist was labeled with (3)H. The bone-adsorption of the (14)C pro-drug following an IV bolus was about 10% compared to 7.8% for the tritiated pro-drug. The difference in release half-life (5.2 and 19.7 days from (3)H and (14)C experiments, respectively) indicated that, after binding to bone, the initial hydrolysis occurred at the ester moiety of the linker releasing the EP4 agonist. The conjugate was found to concentrate in more porous, high-surface-area regions of the long bones. Both (3)H and (14)C experiments indicated a short circulating half-life (1-2 h) in blood.


Asunto(s)
Conservadores de la Densidad Ósea/farmacocinética , Difosfonatos/farmacocinética , Osteoporosis/tratamiento farmacológico , Profármacos/farmacocinética , Subtipo EP4 de Receptores de Prostaglandina E/agonistas , Animales , Conservadores de la Densidad Ósea/química , Huesos/metabolismo , Difosfonatos/química , Femenino , Semivida , Profármacos/química , Ratas , Ratas Sprague-Dawley
5.
Bioorg Med Chem ; 23(13): 3237-47, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-25979376

RESUMEN

An efficient assay for monitoring the activity of the key autophagy-initiating enzyme ATG4B based on a small peptide substrate has been developed. A number of putative small fluorogenic peptide substrates were prepared and evaluated and optimized compounds showed reasonable rates of cleavage but required high enzyme concentrations which limited their value. A modified peptide substrate incorporating a less sterically demanding self-immolative element was designed and synthesized and was shown to have enhanced properties useful for evaluating inhibitors of ATG4B. Substrate cleavage was readily monitored and was linear for up to 4h but enzyme concentrations of about ten-fold higher were required compared to assays using protein substrate LC3 or analogs thereof (such as FRET-LC3). Several known inhibitors of ATG4B were evaluated using the small peptide substrate and gave IC50 values 3-7 fold higher than previously obtained values using the FRET-LC3 substrate.


Asunto(s)
Bioensayo , Cisteína Endopeptidasas/química , Inhibidores de Cisteína Proteinasa/química , Colorantes Fluorescentes/síntesis química , Péptidos/síntesis química , Proteínas Recombinantes de Fusión/química , Secuencia de Aminoácidos , Autofagia , Proteínas Relacionadas con la Autofagia , Cisteína Endopeptidasas/genética , Transferencia Resonante de Energía de Fluorescencia , Colorantes Fluorescentes/química , Humanos , Proteínas Asociadas a Microtúbulos/química , Datos de Secuencia Molecular , Péptidos/química , Proteolisis , Proteínas Recombinantes de Fusión/genética
6.
Bioorg Med Chem ; 22(5): 1708-25, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24508307

RESUMEN

A novel series of bis-indoles derived from naturally occurring marine alkaloid 4 were synthesized and evaluated as inhibitors of methicillin-resistant Staphylococcus aureus (MRSA) pyruvate kinase (PK). PK is not only critical for bacterial survival which would make it a target for development of novel antibiotics, but it is reported to be one of the most highly connected 'hub proteins' in MRSA, and thus should be very sensitive to mutations and making it difficult for the bacteria to develop resistance. From the co-crystal structure of cis-3-4-dihydrohamacanthin B (4) bound to S. aureus PK we were able to identify the pharmacophore needed for activity. Consequently, we prepared simple direct linked bis-indoles such as 10b that have similar anti-MRSA activity as compound 4. Structure-activity relationship (SAR) studies were carried out on 10b and led us to discover more potent compounds such as 10c, 10d, 10k and 10 m with enzyme inhibiting activities in the low nanomolar range that effectively inhibited the bacteria growth in culture with minimum inhibitory concentrations (MIC) for MRSA as low as 0.5 µg/ml. Some potent PK inhibitors, such as 10b, exhibited attenuated antibacterial activity and were found to be substrates for an efflux mechanism in S. aureus. Studies comparing a wild type S. aureus with a construct (S. aureus LAC Δpyk::Erm(R)) that lacks PK activity confirmed that bactericidal activity of 10d was PK-dependant.


Asunto(s)
Staphylococcus aureus Resistente a Meticilina/química , Piruvato Quinasa/antagonistas & inhibidores , Piruvato Quinasa/uso terapéutico , Humanos , Staphylococcus aureus Resistente a Meticilina/aislamiento & purificación , Estructura Molecular , Infecciones Estafilocócicas/microbiología , Relación Estructura-Actividad
7.
Emerg Microbes Infect ; 12(2): 2246594, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37555275

RESUMEN

Antivirals with broad coronavirus activity are important for treating high-risk individuals exposed to the constantly evolving SARS-CoV-2 variants of concern (VOCs) as well as emerging drug-resistant variants. We developed and characterized a novel class of active-site-directed 3-chymotrypsin-like protease (3CLpro) inhibitors (C2-C5a). Our lead direct-acting antiviral (DAA), C5a, is a non-covalent, non-peptide with a dissociation constant of 170 nM against recombinant SARS-CoV-2 3CLpro. The compounds C2-C5a exhibit broad-spectrum activity against Omicron subvariants (BA.5, BQ.1.1, and XBB.1.5) and seasonal human coronavirus-229E infection in human cells. Notably, C5a has median effective concentrations of 30-50 nM against BQ.1.1 and XBB.1.5 in two different human cell lines. X-ray crystallography has confirmed the unique binding modes of C2-C5a to the 3CLpro, which can limit virus cross-resistance to emerging Paxlovid-resistant variants. We tested the effect of C5a with two of our newly discovered host-directed antivirals (HDAs): N-0385, a TMPRSS2 inhibitor, and bafilomycin D (BafD), a human vacuolar H+-ATPase [V-ATPase] inhibitor. We demonstrated a synergistic action of C5a in combination with N-0385 and BafD against Omicron BA.5 infection in human Calu-3 lung cells. Our findings underscore that a SARS-CoV-2 multi-targeted treatment for circulating Omicron subvariants based on DAAs (C5a) and HDAs (N-0385 or BafD) can lead to therapeutic benefits by enhancing treatment efficacy. Furthermore, the high-resolution structures of SARS-CoV-2 3CLpro in complex with C2-C5a will facilitate future rational optimization of our novel broad-spectrum active-site-directed 3C-like protease inhibitors.


Asunto(s)
COVID-19 , Hepatitis C Crónica , Humanos , Inhibidores de Proteasas/farmacología , Antivirales/farmacología , SARS-CoV-2
8.
J Biol Chem ; 286(9): 7290-300, 2011 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-21193398

RESUMEN

Autophagy enables cells to degrade and recycle cytoplasmic materials both as a housekeeping mechanism and in response to extracellular stress such as nutrient deprivation. Recent studies indicate that autophagy also functions as a protective mechanism in response to several cancer therapy agents, making it a prospective therapeutic target. Few pharmacological inhibitors suitable for testing the therapeutic potential of autophagy inhibition in vivo are known. An automated microscopy assay was used to screen >3,500 drugs and pharmacological agents and identified one drug, verteporfin, as an inhibitor of autophagosome accumulation. Verteporfin is a benzoporphyrin derivative used in photodynamic therapy, but it inhibits autophagy without light activation. Verteporfin did not inhibit LC3/Atg8 processing or membrane recruitment in response to autophagic stimuli, but it inhibited drug- and starvation-induced autophagic degradation and the sequestration of cytoplasmic materials into autophagosomes. Transient exposure to verteporfin in starvation conditions reduced cell viability whereas cells in nutrient-rich medium were unaffected by drug treatment. Analysis of structural analogs indicated that the activity of verteporfin requires the presence of a substituted cyclohexadiene at ring A of the porphyrin core but that it can tolerate a number of large substituents at rings C and D. The existence of an autophagy inhibitor among FDA-approved drugs should facilitate the investigation of the therapeutic potential of autophagy inhibition in vivo.


Asunto(s)
Antineoplásicos/farmacología , Autofagia/efectos de los fármacos , Fagosomas/efectos de los fármacos , Porfirinas/farmacología , Antineoplásicos/química , Autofagia/fisiología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Dextranos/farmacocinética , Diseño de Fármacos , Femenino , Fluoresceína-5-Isotiocianato/análogos & derivados , Fluoresceína-5-Isotiocianato/farmacocinética , Proteínas Fluorescentes Verdes/farmacocinética , Humanos , Microscopía Electrónica , Fagosomas/fisiología , Fagosomas/ultraestructura , Porfirinas/química , Verteporfina
9.
Bioorg Med Chem ; 20(6): 2131-40, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22341574

RESUMEN

There is an important medical need for effective therapies to redress the general bone loss associated with advanced osteoporosis. Prostaglandin E(2) and related EP4 receptor agonists have been shown to stimulate bone regrowth but their use has been limited by systemic side effects. Herein is described the design and synthesis of novel dual-action bone-targeting conjugate pro-drugs where two classes of active agents, a bone growth stimulating prostaglandin E(2) EP4 receptor subtype agonist (5 or 6) and a bone resorption inhibitor bisphosphonate, alendronic acid (1), are coupled using metabolically labile carbamate or 4-hydroxyphenylacetic acid based linkers. Radiolabelled conjugates 9, 11a/b and 25 were synthesized and evaluated in vivo in rats for uptake of the conjugate into bone and subsequent release of the EP4 agonists over time. While conjugate 11a/b was taken up (9.0% of initial dose) but not released over two weeks, conjugates 9 and 25 were absorbed at 9.4% and 5.9% uptake of the initial dose and slowly released with half-lives of approximately 2 weeks and 5 days respectively. These conjugates were well tolerated and offer potential for sustained release and dual synergistic activity through their selective bone targeting and local release of the complimentary active components.


Asunto(s)
Alendronato/análogos & derivados , Alendronato/farmacología , Conservadores de la Densidad Ósea/química , Conservadores de la Densidad Ósea/farmacología , Profármacos/química , Profármacos/farmacología , Subtipo EP4 de Receptores de Prostaglandina E/agonistas , Alendronato/síntesis química , Alendronato/farmacocinética , Animales , Conservadores de la Densidad Ósea/síntesis química , Conservadores de la Densidad Ósea/farmacocinética , Huesos/efectos de los fármacos , Huesos/metabolismo , Diseño de Fármacos , Femenino , Osteoporosis/tratamiento farmacológico , Osteoporosis/metabolismo , Fenilacetatos/química , Profármacos/síntesis química , Profármacos/farmacocinética , Ratas , Ratas Sprague-Dawley , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo
10.
Bioorg Med Chem ; 20(24): 7069-82, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23141418

RESUMEN

A novel series of hydrazones were synthesized and evaluated as inhibitors of methicillin-resistant Staphylococcus aureus (MRSA) pyruvate kinase (PK). PK has been identified as one of the most highly connected 'hub proteins' in MRSA. PK has been shown to be critical for bacterial survival which makes it a potential target for development of novel antibiotics and the high degree of connectivity implies it should be very sensitive to mutations and thus less able to develop resistance. PK is not unique to bacteria and thus a critical requirement for such a PK inhibitor would be that it does not inhibit the homologous human enzyme(s) at therapeutic concentrations. Several MRSA PK inhibitors (including 8d) were identified using in silico screening combined with enzyme assays and were found to be selective for bacterial enzyme compared to four human PK isoforms (M1, M2, R and L). However these lead compounds did not show significant inhibitory activity for MRSA growth presumably due to poor bacterial cell penetration. Structure-activity relationship (SAR) studies were carried out on 8d and led us to discover more potent compounds with enzyme inhibiting activities in the low nanomolar range and some were found to effectively inhibit bacteria growth in culture with minimum inhibitory concentrations (MIC) as low as 1 µg/mL. These inhibitors bind in two elongated flat clefts found at the minor interfaces in the homo-tetrameric enzyme complex and the observed SAR is in keeping with the size and electronic constraints of these binding sites. Access to the corresponding sites in the human enzyme is blocked.


Asunto(s)
Antiinfecciosos/química , Antiinfecciosos/farmacología , Staphylococcus aureus Resistente a Meticilina/enzimología , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Piruvato Quinasa/antagonistas & inhibidores , Humanos , Modelos Moleculares , Piruvato Quinasa/metabolismo , Relación Estructura-Actividad
11.
STAR Protoc ; 3(2): 101258, 2022 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-35434660

RESUMEN

We describe a protocol to identify the binding site(s) for a drug called ivacaftor that potentiates the CFTR chloride channel. We use photoaffinity probes-based on the structure of ivacaftor-to covalently modify the CFTR protein at the region that constitutes the drug binding site(s). We define the methods for photo-labeling CFTR, its membrane extraction, and enzymatic digestion using trypsin. We then describe the experimental methods to identify the modified peptides by using mass spectrometry. For complete details on the use and execution of this protocol, please refer to Laselva et al. (2021).


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística , Fibrosis Quística , Sitios de Unión , Fibrosis Quística/tratamiento farmacológico , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Humanos , Transporte Iónico , Mutación
12.
Antimicrob Agents Chemother ; 55(5): 2042-53, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21357306

RESUMEN

Novel classes of antimicrobials are needed to address the challenge of multidrug-resistant bacteria such as methicillin-resistant Staphylococcus aureus (MRSA). Using the architecture of the MRSA interactome, we identified pyruvate kinase (PK) as a potential novel drug target based upon it being a highly connected, essential hub in the MRSA interactome. Structural modeling, including X-ray crystallography, revealed discrete features of PK in MRSA, which appeared suitable for the selective targeting of the bacterial enzyme. In silico library screening combined with functional enzymatic assays identified an acyl hydrazone-based compound (IS-130) as a potent MRSA PK inhibitor (50% inhibitory concentration [IC50] of 0.1 µM) with >1,000-fold selectivity over human PK isoforms. Medicinal chemistry around the IS-130 scaffold identified analogs that more potently and selectively inhibited MRSA PK enzymatic activity and S. aureus growth in vitro (MIC of 1 to 5 µg/ml). These novel anti-PK compounds were found to possess antistaphylococcal activity, including both MRSA and multidrug-resistant S. aureus (MDRSA) strains. These compounds also exhibited exceptional antibacterial activities against other Gram-positive genera, including enterococci and streptococci. PK lead compounds were found to be noncompetitive inhibitors and were bactericidal. In addition, mutants with significant increases in MICs were not isolated after 25 bacterial passages in culture, indicating that resistance may be slow to emerge. These findings validate the principles of network science as a powerful approach to identify novel antibacterial drug targets. They also provide a proof of principle, based upon PK in MRSA, for a research platform aimed at discovering and optimizing selective inhibitors of novel bacterial targets where human orthologs exist, as leads for anti-infective drug development.


Asunto(s)
Antibacterianos/farmacología , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Staphylococcus aureus Resistente a Meticilina/enzimología , Piruvato Quinasa/metabolismo , Secuencia de Aminoácidos , Electroforesis en Gel de Poliacrilamida , Humanos , Staphylococcus aureus Resistente a Meticilina/genética , Datos de Secuencia Molecular , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Piruvato Quinasa/química , Piruvato Quinasa/genética , Homología de Secuencia de Aminoácido
13.
Bioorg Med Chem Lett ; 21(3): 920-3, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21232956

RESUMEN

The trifluoroethylamine group found in cathepsin K inhibitors like odanacatib can be replaced by a difluoroethylamine group. This change increased the basicity of the nitrogen which positively impacted the log D. This translated into an improved oral bioavailability in pre-clinical species. Difluoroethylamine compounds exhibit a similar potency against cathepsin K and selectivity profile against other cathepsins when compared to trifluoroethylamine analogs.


Asunto(s)
Catepsina K/antagonistas & inhibidores , Etilaminas/química , Inhibidores de Proteasas/química , Administración Oral , Amidas/química , Animales , Compuestos de Bifenilo/química , Compuestos de Bifenilo/farmacología , Catepsina K/metabolismo , Perros , Etilaminas/síntesis química , Etilaminas/farmacocinética , Inhibidores de Proteasas/síntesis química , Inhibidores de Proteasas/farmacocinética , Ratas
14.
ACS Pharmacol Transl Sci ; 4(2): 908-925, 2021 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-33860210

RESUMEN

A series of bone-targeting EP4 receptor agonist conjugate prodrugs were prepared wherein a potent EP4 receptor agonist was bound to a biologically inactive, bisphosphonate-based bone-targeting moiety. Singly and doubly radiolabeled conjugates were synthesized and were shown to be stable in blood, to be rapidly eliminated from the bloodstream, and to be effectively taken up into bone in vivo after intravenous dosing. From these preliminary studies a preferred conjugate 4 (also known as C3 and Mes-1007) was selected for follow up biodistribution and elimination studies. Doubly radiolabeled conjugate 4 was found to partition largely to the liver and bones, and both labels were eliminated from liver at the same rate indicating the conjugate was eliminated intact. Quantification of the labels in bones indicated that free EP4 agonist (EP4a)(2a) was released from bone-bound 4 with a half-time of about 7 days. When dosed orally, radiolabeled 4 was not absorbed and passed through the gastrointestinal tract essentially unchanged, and only traces of radiolabeled 4 were found in the liver, blood, or bones. 4 was found to bind rapidly and completely to powdered bone mineral or to various forms of calcium phosphate, forming a stable matrix suitable for implant and that could made into powders or solid forms and be sterilized without decomposition or release of 4. Basic hydrolysis released free EP4 agonist 2a quantitatively from the material.

15.
Bone ; 145: 115867, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33524637

RESUMEN

OBJECTIVE: Osteogenesis imperfecta (OI) is a heritable bone fragility disorder that is usually caused by mutations affecting collagen type I synthesis in osteoblasts. Bisphosphonates are widely used to decrease fracture rate but are only partially effective. Bone anabolic compounds, such as prostaglandin E2 receptor 4 (EP4) agonists may be an alternative treatment approach. Here we assessed the effect of Mes-1007, a novel bone-targeted EP4 agonist in Jrt mice, a model of severe OI. STUDY DESIGN: Experimental study. RESULTS: Male 8-week old wild type (WT) and OI mice were randomly assigned to 4 weeks of three intraperitoneal injections per week with Mes-1007 (25 mg per kg body mass), phosphate-buffered saline, zoledronate (5 µg per kg), or a combination treatment of zoledronate and Mes-1007. Treatment with Mes-1007 alone did not lead to higher trabecular bone volume per tissue volume (BV/TV) in the distal femur or lumbar vertebra 4 in either WT or OI mice. Treatment with zoledronate alone was associated with a significant increase in distal femur and vertebra BV/TV in both genotypes. In zoledronate-treated WT and OI mice, Mes-1007 increased bone formation rate in vertebral trabecular bone and had an additive effect on BV/TV. Vertebral BV/TV in OI mice that received zoledronate or Mes-1007/zoledronate combination treatment was similar to untreated WT mice (p = 0.25). At the femoral midshaft, Mes-1007/zoledronate combination treatment increased cortical thickness in both genotypes and led to higher periosteal diameter in OI mice. Three-point bending tests of femurs showed that Mes-1007/zoledronate combination treatment increased the stiffness, load at yield and maximal load in WT but not in OI mice. CONCLUSION: Dosing Mes-1007 in combination with zoledronate improved the bone properties in a manner that is consistent with a mechanism of action of EP4 agonists on bone and additive to effects of anti-resorptives typified by zoledronate.


Asunto(s)
Osteogénesis Imperfecta , Animales , Densidad Ósea , Huesos , Dinoprostona , Modelos Animales de Enfermedad , Fémur , Masculino , Ratones , Osteogénesis , Osteogénesis Imperfecta/tratamiento farmacológico
16.
J Med Chem ; 64(20): 14968-14982, 2021 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-34661404

RESUMEN

Prostate cancer (PCa) patients undergoing androgen deprivation therapy almost invariably develop castration-resistant prostate cancer (CRPC). Targeting the androgen receptor (AR) Binding Function-3 (BF3) site offers a promising option to treat CRPC. However, BF3 inhibitors have been limited by poor potency or inadequate metabolic stability. Through extensive medicinal chemistry, molecular modeling, and biochemistry, we identified 2-(5,6,7-trifluoro-1H-Indol-3-yl)-quinoline-5-carboxamide (VPC-13789), a potent AR BF3 antagonist with markedly improved pharmacokinetic properties. We demonstrate that VPC-13789 suppresses AR-mediated transcription, chromatin binding, and recruitment of coregulatory proteins. This novel AR antagonist selectively reduces the growth of both androgen-dependent and enzalutamide-resistant PCa cell lines. Having demonstrated in vitro efficacy, we developed an orally bioavailable prodrug that reduced PSA production and tumor volume in animal models of CRPC with no observed toxicity. VPC-13789 is a potent, selective, and orally bioavailable antiandrogen with a distinct mode of action that has a potential as novel CRPC therapeutics.


Asunto(s)
Antagonistas de Andrógenos/farmacología , Antineoplásicos/farmacología , Desarrollo de Medicamentos , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Quinolinas/farmacología , Receptores Androgénicos/metabolismo , Administración Oral , Antagonistas de Andrógenos/administración & dosificación , Antagonistas de Andrógenos/química , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Disponibilidad Biológica , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Masculino , Modelos Moleculares , Estructura Molecular , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/patología , Quinolinas/administración & dosificación , Quinolinas/química , Relación Estructura-Actividad
17.
Nat Cell Biol ; 23(7): 796-807, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34239062

RESUMEN

Inflammatory bowel diseases present with elevated levels of intestinal epithelial cell (IEC) death, which compromises the gut barrier, activating immune cells and triggering more IEC death. The endogenous signals that prevent IEC death and break this vicious cycle, allowing resolution of intestinal inflammation, remain largely unknown. Here we show that prostaglandin E2 signalling via the E-type prostanoid receptor 4 (EP4) on IECs represses epithelial necroptosis and induces resolution of colitis. We found that EP4 expression correlates with an improved IBD outcome and that EP4 activation induces a transcriptional signature consistent with resolution of intestinal inflammation. We further show that dysregulated necroptosis prevents resolution, and EP4 agonism suppresses necroptosis in human and mouse IECs. Mechanistically, EP4 signalling on IECs converges on receptor-interacting protein kinase 1 to suppress tumour necrosis factor-induced activation and membrane translocation of the necroptosis effector mixed-lineage kinase domain-like pseudokinase. In summary, our study indicates that EP4 promotes the resolution of colitis by suppressing IEC necroptosis.


Asunto(s)
Colitis/metabolismo , Colon/metabolismo , Dinoprostona/metabolismo , Células Epiteliales/metabolismo , Mucosa Intestinal/metabolismo , Necroptosis , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo , Animales , Antiinflamatorios/farmacología , Colitis/inducido químicamente , Colitis/patología , Colitis/prevención & control , Colon/efectos de los fármacos , Colon/patología , Sulfato de Dextran , Modelos Animales de Enfermedad , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Células HT29 , Humanos , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Quinasas Quinasa Quinasa PAM/genética , Quinasas Quinasa Quinasa PAM/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Necroptosis/efectos de los fármacos , Organoides , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Subtipo EP4 de Receptores de Prostaglandina E/agonistas , Subtipo EP4 de Receptores de Prostaglandina E/genética , Transducción de Señal
18.
Bioorg Med Chem Lett ; 20(18): 5502-5, 2010 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-20709547

RESUMEN

The SAR study of a series of 6-aryloxymethyl-8-aryl substituted quinolines is described. Optimization of the series led to the discovery of compound 26b, a highly potent (IC50=0.6 nM) and selective PDE4D inhibitor with a 75-fold selectivity over the A, B, and C subtypes and over 18,000-fold selectivity against other PDE family members. Rat pharmacokinetics and tissue distribution are also summarized.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/metabolismo , Inhibidores de Fosfodiesterasa/química , Inhibidores de Fosfodiesterasa/farmacología , Quinolinas/química , Quinolinas/farmacología , Animales , Asma/tratamiento farmacológico , Humanos , Concentración 50 Inhibidora , Masculino , Inhibidores de Fosfodiesterasa/síntesis química , Inhibidores de Fosfodiesterasa/farmacocinética , Quinolinas/síntesis química , Quinolinas/farmacocinética , Ratas , Ratas Wistar , Relación Estructura-Actividad
19.
Bioorg Med Chem Lett ; 20(22): 6387-93, 2010 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-20933411

RESUMEN

The structure-activity relationship of a novel series of 8-biarylnaphthyridinones acting as type 4 phosphodiesterase (PDE4) inhibitors for the treatment of long-term memory loss and mild cognitive impairment is described herein. The manuscript describes a new paradigm for the development of PDE4 inhibitor targeting CNS indications. This effort led to the discovery of the clinical candidate MK-0952, an intrinsically potent inhibitor (IC(50)=0.6 nM) displaying limited whole blood activity (IC(50)=555 nM). Supporting in vivo results in two preclinical efficacy tests and one test assessing adverse effects are also reported. The comparative profiles of MK-0952 and two other Merck compounds are described to validate the proposed hypothesis.


Asunto(s)
Trastornos del Conocimiento/tratamiento farmacológico , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/efectos de los fármacos , Ciclopropanos/farmacología , Compuestos Heterocíclicos con 2 Anillos/farmacología , Memoria a Largo Plazo/efectos de los fármacos , Inhibidores de Fosfodiesterasa/farmacología , Animales , Ciclopropanos/química , Ciclopropanos/uso terapéutico , Perros , Femenino , Compuestos Heterocíclicos con 2 Anillos/química , Compuestos Heterocíclicos con 2 Anillos/uso terapéutico , Humanos , Macaca mulatta , Masculino , Inhibidores de Fosfodiesterasa/química , Inhibidores de Fosfodiesterasa/uso terapéutico , Ratas , Relación Estructura-Actividad
20.
Bioorg Med Chem Lett ; 20(3): 887-92, 2010 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-20061146

RESUMEN

MK-0674 is a potent and selective cathepsin K inhibitor from the same structural class as odanacatib with a comparable inhibitory potency profile against Cat K. It is orally bioavailable and exhibits long half-life in pre-clinical species. In vivo studies using deuterated MK-0674 show stereoselective epimerization of the alcohol stereocenter via an oxidation/reduction cycle. From in vitro incubations, two metabolites could be identified: the hydroxyleucine and the glucuronide conjugate which were confirmed using authentic synthetic standards.


Asunto(s)
Compuestos de Bifenilo/administración & dosificación , Compuestos de Bifenilo/farmacocinética , Catepsina K/antagonistas & inhibidores , Inhibidores de Cisteína Proteinasa/administración & dosificación , Inhibidores de Cisteína Proteinasa/farmacocinética , Descubrimiento de Drogas/métodos , Administración Oral , Animales , Disponibilidad Biológica , Compuestos de Bifenilo/química , Catepsina K/metabolismo , Inhibidores de Cisteína Proteinasa/química , Perros , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Macaca mulatta , Conejos , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA