Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Acta Biomater ; 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38702010

RESUMEN

Cascade-reaction containers generating reactive oxygen species (ROS) as an alternative for antibiotic-based strategies for bacterial infection control, require endogenous oxygen-sources and ROS-generation close to or preferably inside target bacteria. Here, this is achieved by cetyltrimethylammonium-chloride (CTAC) assisted in situ metabolic labeling and incorporation of mesoporous SiO2-nanoparticles, dual-loaded with glucose-oxidase and Fe3O4-nanoparticles as cascade-reaction containers, inside bacterial cell walls. First, azide-functionalized d-alanine (D-Ala-N3) was inserted in cell wall peptidoglycan layers of growing Gram-positive pathogens. In Gram-negatives, this could only be achieved after outer lipid-membrane permeabilization, using a low concentration of CTAC. Low concentrations of CTAC had no adverse effect on in vitro blood clotting or hemolysis nor on the health of mice when blood-injected. Next, dibenzocyclooctyne-polyethylene-glycol modified, SiO2-nanoparticles were in situ click-reacted with d-Ala-N3 in bacterial cell wall peptidoglycan layers. Herewith, a two-step cascade-reaction is facilitated inside bacteria, in which glucose-oxidase generates H2O2 at endogenously-available glucose concentrations, while subsequently Fe3O4-nanoparticles catalyze generation of •OH from the H2O2 generated. Generation of •OH inside bacterial cell walls by dual-loaded mesoporous SiO2-nanoparticles yielded more effective in vitro killing of both planktonic Gram-positive and Gram-negative bacteria suspended in 10 % plasma than SiO2-nanoparticles solely loaded with glucose-oxidase. Gram-positive or Gram-negative bacterially induced sepsis in mice could be effectively treated by in situ pre-treatment with tail-vein injected CTAC and d-Ala-N3, followed by injection of dual-loaded cascade-reaction containers without using antibiotics. This makes in situ metabolic incorporation of cascade-reaction containers as described attractive for further investigation with respect to the control of other types of infections comprising planktonic bacteria. STATEMENT OF SIGNIFICANCE: In situ metabolic-incorporation of cascade-reaction-containers loaded with glucose-oxidase and Fe3O4 nanoparticles into bacterial cell-wall peptidoglycan is described, yielding ROS-generation from endogenous glucose, non-antibiotically killing bacteria before ROS inactivates. Hitherto, only Gram-positives could be metabolically-labeled, because Gram-negatives possess two lipid-membranes. The outer membrane impedes direct access to the peptidoglycan. This problem was solved by outer-membrane permeabilization using a quaternary-ammonium compound. Several studies on metabolic-labeling perform crucial labeling steps during bacterial-culturing that in real-life should be part of a treatment. In situ metabolic-incorporation as described, can be applied in well-plates during in vitro experiments or in the body as during in vivo animal experiments. Surprisingly, metabolic-incorporation proceeded unhampered in blood and a murine, bacterially-induced sepsis could be well treated.

2.
Biomaterials ; 308: 122576, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38640785

RESUMEN

Biomaterial-associated infection (BAI) is considered a unique infection due to the presence of a biomaterial yielding frustrated immune-cells, ineffective in clearing local micro-organisms. The involvement of surface-adherent/surface-adapted micro-organisms in BAI, logically points to biomaterial surface-modifications for BAI-control. Biomaterial surface-modification is most suitable for prevention before adhering bacteria have grown into a mature biofilm, while BAI-treatment is virtually impossible through surface-modification. Hundreds of different surface-modifications have been proposed for BAI-control but few have passed clinical trials due to the statistical near-impossibility of benefit-demonstration. Yet, no biomaterial surface-modification forwarded, is clinically embraced. Collectively, this leads us to conclude that surface-modification is a dead-end road. Accepting that BAI is, like most human infections, due to surface-adherent biofilms (though not always to a foreign material), and regarding BAI as a common infection, opens a more-generally-applicable and therewith easier-to-validate road. Pre-clinical models have shown that stimuli-responsive nano-antimicrobials and antibiotic-loaded nanocarriers exhibit prolonged blood-circulation times and can respond to a biofilm's micro-environment to penetrate and accumulate within biofilms, prompt ROS-generation and synergistic killing with antibiotics of antibiotic-resistant pathogens without inducing further antimicrobial-resistance. Moreover, they can boost frustrated immune-cells around a biomaterial reducing the importance of this unique BAI-feature. Time to start exploring the nano-road for BAI-control.


Asunto(s)
Materiales Biocompatibles , Biopelículas , Nanotecnología , Propiedades de Superficie , Humanos , Materiales Biocompatibles/química , Biopelículas/efectos de los fármacos , Nanotecnología/métodos , Animales , Infecciones Relacionadas con Prótesis/prevención & control , Prótesis e Implantes , Antibacterianos/farmacología , Antibacterianos/uso terapéutico
3.
Molecules ; 27(22)2022 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-36432026

RESUMEN

(1) Background: Nuclear factor κB (NF-κB) is an important transcriptional regulator that regulates the inflammatory pathway and plays a key role in cellular inflammatory and immune responses. The presence of a high concentration of NF-κB is positively correlated with the severity of inflammation. Therefore, the inhibition of this pathway is an important therapeutic target for the treatment of various types of inflammation; (2) Methods: we designed and synthesized 23 mollugin derivatives and evaluated their inhibitory activity against NF-κB transcription; (3) Results: Compound 6d exhibited the most promising inhibitory activity (IC50 = 3.81 µM) and did not show any significant cytotoxicity against the tested cell lines. Investigation of the mechanism of action indicated that 6d down-regulated NF-κB expression, possibly by suppressing TNF-α-induced expression of the p65 protein. Most of the compounds exhibited potent anti-inflammatory activity. Compound 4f was the most potent compound with 83.08% inhibition of inflammation after intraperitoneal administration, which was more potent than mollugin and the reference drugs (ibuprofen and mesalazine). ADMET prediction analysis indicated that compounds 6d and 4f had good pharmacokinetics and drug-like behavior; (4) Conclusions: Several series of mollugin derivatives were designed, synthesized, and evaluated for NF-κB inhibitory activity and toxicity. These results provide an initial basis for the development of 4f and 6d as potential anti-inflammatory agents.


Asunto(s)
FN-kappa B , Piranos , Humanos , Inflamación , Inyecciones Intraperitoneales
4.
J Control Release ; 352: 460-471, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36341930

RESUMEN

Exposure of infectious biofilms to dispersants induces high bacterial concentrations in blood that may cause sepsis. Preventing sepsis requires simultaneous biofilm dispersal and bacterial killing. Here, self-targeting DCPA(2-(4-((1,5-bis(octadecenoyl)1,5-dioxopentan-2-yl)carbamoyl)pyridin-1-ium-1-yl)acetate) liposomes with complexed water were self-assembled with ciprofloxacin loaded in-membrane and PEGylated as a lipid-membrane component, together with bromelain loaded in-core. Inside biofilms, DCPA-H2O and PEGylated ciprofloxacin became protonated, disturbing the balance in the lipid-membrane to cause liposome-burst and simultaneous release of bromelain and ciprofloxacin. Simultaneous release of bromelain and ciprofloxacin enhanced bacterial killing in Staphylococcus aureus biofilms as compared with free bromelain and/or ciprofloxacin. After tail-vein injection in mice, liposomes accumulated inside intra-abdominal staphylococcal biofilms. Subsequent liposome-burst and simultaneous release of bromelain and ciprofloxacin yielded degradation of the biofilm matrix by bromelain and higher bacterial killing without inducing septic symptoms as obtained by injection of free bromelain and ciprofloxacin. This shows the advantage of simultaneous release from liposomes of bromelain and ciprofloxacin inside a biofilm.


Asunto(s)
Bromelaínas , Sepsis , Animales , Ratones , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Biopelículas , Ciprofloxacina/farmacología , Lípidos , Liposomas , Pruebas de Sensibilidad Microbiana , Polietilenglicoles , Protones , Sepsis/tratamiento farmacológico
5.
Biomater Sci ; 10(13): 3575-3584, 2022 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-35635244

RESUMEN

Protein therapeutics have been viewed as powerful candidates for cancer treatment by virtue of highly specific bioactivity and minimized adverse effects. However, the intracellular delivery of protein drugs remains enormously challenging due to multiple successive biological barriers in vivo. Herein, a bioinspired nanochaperone is developed to assist proteins in vanquishing the sequential physiological barriers in a holistic manner and enhance synergistic tumor therapy. By concurrently mimicking the N-terminal-binding domain and C-terminal-stabilizing domain of natural chaperones, this nanochaperone can efficiently capture the protein by multiple interactions and hide them in the confined spaces on the surface, serving as a shield to resist enzymatic degradation and avoid immune clearance during blood circulation. Upon reaching the tumor site, the nanochaperone rapidly responds to the acidic tumor microenvironment and turns into partial protonation, acting as a spear to facilitate tumor cellular internalization. More importantly, further protonation of nanochaperone in the lysosome of tumor cells enables it to blast the lysosome and achieve cytosolic protein delivery with reserved bioactivities. Furthermore, this nanochaperone-based protein transduction strategy is demonstrated to combine with small-molecule drugs to synergistically amplify the anti-tumor therapeutic effect in vitro and in vivo, providing a potential platform for the exploitation of diverse combinations of anti-tumor therapies.


Asunto(s)
Antineoplásicos , Neoplasias , Antineoplásicos/química , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Microambiente Tumoral
6.
Bioact Mater ; 14: 321-334, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35386819

RESUMEN

Cascade-reaction chemistry can generate reactive-oxygen-species that can be used for the eradication of infectious biofilms. However, suitable and sufficient oxygen sources are not always available near an infection site, while the reactive-oxygen-species generated are short-lived. Therefore, we developed a magnetic cascade-reaction container composed of mesoporous Fe3O4@SiO2 nanoparticles containing glucose-oxidase and l-arginine for generation of reactive-oxygen-species. Glucose-oxidase was conjugated with APTES facilitating coupling to Fe3O4@SiO2 nanoparticles and generation of H2O2 from glucose. l-arginine was loaded into the nanoparticles to generate NO from the H2O2 generated. Using an externally-applied magnetic field, cascade-reaction containers could be homogeneously distributed across the depth of an infectious biofilm. Cascade-reaction containers with coupled glucose-oxidase were effective in killing planktonic, Gram-positive and Gram-negative bacteria. Additional efficacy of the l-arginine based second cascade-reaction was only observed when H2O2 as well as NO were generated in-biofilm. In vivo accumulation of cascade-reaction containers inside abdominal Staphylococcus aureus biofilms upon magnetic targeting was observed real-time in living mice through an implanted, intra-vital window. Moreover, vancomycin-resistant, abdominal S. aureus biofilms could be eradicated consuming solely endogenous glucose, without any glucose addition. Herewith, a new, non-antibiotic-based infection-control strategy has been provided, constituting a welcome addendum to the shrinking clinical armamentarium to control antibiotic-resistant bacterial infections.

7.
J Mater Chem B ; 10(14): 2316-2322, 2022 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-35129564

RESUMEN

Self-targeting antimicrobial platforms have yielded new possibilities for the treatment of infectious biofilms. Self-targeting involves stealth transport through the blood circulation towards an infectious biofilm, where the antimicrobial platform penetrates and accumulates in a biofilm in response to a change in environmental conditions, such as local pH. In a final step, nano-antimicrobials need to be activated or the antimicrobial cargo of nanocarriers released. Zwitterions possess both cationic and anionic groups, allowing full reversal in zeta potential from below to above zero in response to a change in environmental conditions. Electrolyte-based platforms generally do not have the ability to change their zeta potentials from below to above zero. Zwitterions for use in self-targeting platforms are usually hydrophilic and have a negative charge under physiological conditions (pH 7.4) providing low adsorption of proteins and assisting blood circulation. However, near or in the acidic environment of a biofilm, they become positively-charged yielding targeting, penetration and accumulation in the biofilm through electrostatic double-layer attraction to negatively-charged bacteria. Response-times to pH changes vary, depending on the way the zwitterion or electrolyte is built in a platform. Self-targeting zwitterion-based platforms with a short response-time in vitro yield different accumulation kinetics in abdominal biofilms in living mice than platforms with a longer response-time. In vivo experiments in mice also proved that self-targeting, pH-responsive zwitterion-based platforms provide a feasible approach for clinical control of bacterial infections. Clinically however, also other conditions than infection may yield an acidic environment. Therefore, it remains to be seen whether pH is a sufficiently unique recognition sign to direct self-targeting platforms to an infectious biofilm or whether (additional) external targeting through e.g. near-infrared irradiation or magnetic field application is needed.


Asunto(s)
Antiinfecciosos , Biopelículas , Animales , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Antiinfecciosos/farmacología , Interacciones Hidrofóbicas e Hidrofílicas , Rayos Infrarrojos , Ratones
8.
Chemistry ; 28(4): e202103114, 2022 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-34820923

RESUMEN

We designed, synthesized, and characterized a tri-block copolymer. Its hydrophobic part, a chain of histone deacetylase inhibitor (HDACi) prodrug, was symmetrically flanked by two identical PEG blocks, whereas the built-in HDACi was a linear molecule, terminated with a thiol at one end, and a hydroxyl group at the other. Such a feature facilitated end-to-end linkage of prodrugs through alternatively aligned disulfides and carbonates. The disulfides served dual roles: redox sensors of smart nanomedicine, and warheads of masked HDACi drugs. This approach, carefully designed to benefit both control-release and efficacy, is conceptually novel for optimizing drug units in nanomedicine. Micelles from this designer polyprodrug released only PEG, CO2 and HDACi, and synergized with DOX against HCT116 cells, demonstrating its widespread potential in combination therapy. Our work highlights, for the first time, the unique advantage of thiol-based drug molecules in nanomedicine design.


Asunto(s)
Inhibidores de Histona Desacetilasas , Profármacos , Doxorrubicina , Micelas , Polietilenglicoles
9.
Angew Chem Int Ed Engl ; 60(32): 17714-17719, 2021 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-34028150

RESUMEN

A lipid named DCPA was synthesized under microwave-assisted heating. DCPA possesses a pyridine betaine, hydrophilic group that can be complexed with water through hydrogen bonding (DCPA-H2 O). DCPA-H2 O liposomes became protonated relatively fast already at pH<6.8, due to the high HOMO binding energy of DCPA-H2 O. In murine models, DCPA-H2 O liposomes had longer blood circulation times than natural DPPC or cationic DCPM liposomes, while after tail-vein injection DCPA-H2 O liposomes targeted faster to solid tumors and intra-abdominal infectious biofilms. Therapeutic efficacy in a murine, infected wound-healing model of tail-vein injected ciprofloxacin-loaded DCPA-H2 O liposomes exceeded the ones of clinically applied ciprofloxacin as well as of ciprofloxacin-loaded DPPC or DCPM liposomes.


Asunto(s)
Portadores de Fármacos/farmacocinética , Liposomas/farmacocinética , Neoplasias/diagnóstico por imagen , Infecciones Estafilocócicas/diagnóstico por imagen , Agua/química , Acetatos/síntesis química , Acetatos/farmacocinética , Animales , Antibacterianos/uso terapéutico , Biopelículas , Ciprofloxacina/uso terapéutico , Portadores de Fármacos/síntesis química , Femenino , Colorantes Fluorescentes/química , Células Hep G2 , Humanos , Concentración de Iones de Hidrógeno , Liposomas/química , Masculino , Ratones Endogámicos BALB C , Mycobacterium tuberculosis/fisiología , Compuestos de Piridinio/síntesis química , Compuestos de Piridinio/farmacocinética , Ratas Sprague-Dawley , Rodaminas/química , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/fisiopatología , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/fisiología , Tuberculosis/diagnóstico por imagen , Tuberculosis/fisiopatología
10.
Bioorg Med Chem Lett ; 30(24): 127652, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33130293

RESUMEN

Hypoxia-inducible factor 1α (HIF-1α) is a known regulator of tumor cell proliferation, migration, and angiogenesis. The presence of a high concentration of HIF-1α is positively correlated with the severity of cancer. Therefore, the inhibition of this pathway represents an important therapeutic target for the treatment of various types of cancer. Here, we designed and synthesized 30 panaxadiol (PD) derivatives and evaluated their inhibitory activities against HIF-1α transcription. Of these, compound 3l exhibited the most promising inhibitory activity (IC50 = 3.7 µM) and showed significantly decreased cytotoxicity compared with PD. Compound 9e exhibited the strongest cytotoxic effect and may be considered for further preclinical development.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Ginsenósidos/química , Ginsenósidos/farmacología , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Antineoplásicos/síntesis química , Línea Celular Tumoral , Ginsenósidos/síntesis química , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Relación Estructura-Actividad , Activación Transcripcional/efectos de los fármacos
11.
Front Chem ; 7: 872, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31998680

RESUMEN

Many nanotechnology-based antimicrobials and antimicrobial-delivery-systems have been developed over the past decades with the aim to provide alternatives to antibiotic treatment of infectious-biofilms across the human body. Antimicrobials can be loaded into nanocarriers to protect them against de-activation, and to reduce their toxicity and potential, harmful side-effects. Moreover, antimicrobial nanocarriers such as micelles, can be equipped with stealth and pH-responsive features that allow self-targeting and accumulation in infectious-biofilms at high concentrations. Micellar and liposomal nanocarriers differ in hydrophilicity of their outer-surface and inner-core. Micelles are self-assembled, spherical core-shell structures composed of single layers of surfactants, with hydrophilic head-groups and hydrophobic tail-groups pointing to the micellar core. Liposomes are composed of lipids, self-assembled into bilayers. The hydrophilic head of the lipids determines the surface properties of liposomes, while the hydrophobic tail, internal to the bilayer, determines the fluidity of liposomal-membranes. Therefore, whereas micelles can only be loaded with hydrophobic antimicrobials, hydrophilic antimicrobials can be encapsulated in the hydrophilic, aqueous core of liposomes and hydrophobic or amphiphilic antimicrobials can be inserted in the phospholipid bilayer. Nanotechnology-derived liposomes can be prepared with diameters <100-200 nm, required to prevent reticulo-endothelial rejection and allow penetration into infectious-biofilms. However, surface-functionalization of liposomes is considerably more difficult than of micelles, which explains while self-targeting, pH-responsive liposomes that find their way through the blood circulation toward infectious-biofilms are still challenging to prepare. Equally, development of liposomes that penetrate over the entire thickness of biofilms to provide deep killing of biofilm inhabitants still provides a challenge. The liposomal phospholipid bilayer easily fuses with bacterial cell membranes to release high antimicrobial-doses directly inside bacteria. Arguably, protection against de-activation of antibiotics in liposomal nanocarriers and their fusogenicity constitute the biggest advantage of liposomal antimicrobial carriers over antimicrobials free in solution. Many Gram-negative and Gram-positive bacterial strains, resistant to specific antibiotics, have been demonstrated to be susceptible to these antibiotics when encapsulated in liposomal nanocarriers. Recently, also progress has been made concerning large-scale production and long-term storage of liposomes. Therewith, the remaining challenges to develop self-targeting liposomes that penetrate, accumulate and kill deeply in infectious-biofilms remain worthwhile to pursue.

12.
Assay Drug Dev Technol ; 11(5): 308-25, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23772552

RESUMEN

Small ubiquitin-like modifier (SUMO) belongs to the family of ubiquitin-like proteins (Ubls) that can be reversibly conjugated to target-specific lysines on substrate proteins. Although covalently sumoylated products are readily detectible in gel-based assays, there has been little progress toward the development of robust quantitative sumoylation assay formats for the evaluation of large compound libraries. In an effort to identify inhibitors of ubiquitin carrier protein 9 (Ubc9)-dependent sumoylation, a high-throughput fluorescence polarization assay was developed, which allows detection of Lys-1201 sumoylation, corresponding to the major site of functional sumoylation within the transcriptional repressor trichorhino-phalangeal syndrome type I protein (TRPS1). A minimal hexapeptide substrate peptide, TMR-VVK1201TEK, was used in this assay format to afford high-throughput screening of the GlaxoSmithKline diversity compound collection. A total of 728 hits were confirmed but no specific noncovalent inhibitors of Ubc9 dependent trans-sumoylation were found. However, several diaminopyrimidine compounds were identified as inhibitors in the assay with IC50 values of 12.5 µM. These were further characterized to be competent substrates which were subject to sumoylation by SUMO-Ubc9 and which were competitive with the sumoylation of the TRPS1 peptide substrates.


Asunto(s)
Proteínas de Unión al ADN/antagonistas & inhibidores , Evaluación Preclínica de Medicamentos/métodos , Mapeo de Interacción de Proteínas/métodos , Espectrometría de Fluorescencia/métodos , Sumoilación/efectos de los fármacos , Factores de Transcripción/antagonistas & inhibidores , Enzimas Ubiquitina-Conjugadoras/antagonistas & inhibidores , Sitios de Unión , Unión Proteica , Proteínas Represoras
13.
Chem Biol ; 18(2): 177-86, 2011 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-21338916

RESUMEN

c-Abl kinase activity is regulated by a unique mechanism involving the formation of an autoinhibited conformation in which the N-terminal myristoyl group binds intramolecularly to the myristoyl binding site on the kinase domain and induces the bending of the αI helix that creates a docking surface for the SH2 domain. Here, we report a small-molecule c-Abl activator, DPH, that displays potent enzymatic and cellular activity in stimulating c-Abl activation. Structural analyses indicate that DPH binds to the myristoyl binding site and prevents the formation of the bent conformation of the αI helix through steric hindrance, a mode of action distinct from the previously identified allosteric c-Abl inhibitor, GNF-2, that also binds to the myristoyl binding site. DPH represents the first cell-permeable, small-molecule tool compound for c-Abl activation.


Asunto(s)
Descubrimiento de Drogas , Hidantoínas/metabolismo , Hidantoínas/farmacología , Proteínas Proto-Oncogénicas c-abl/metabolismo , Pirazoles/metabolismo , Pirazoles/farmacología , Secuencia de Aminoácidos , Sitios de Unión , Cristalografía por Rayos X , Activación Enzimática/efectos de los fármacos , Células Hep G2 , Humanos , Hidantoínas/química , Modelos Moleculares , Datos de Secuencia Molecular , Permeabilidad , Fosforilación/efectos de los fármacos , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-abl/química , Proteínas Proto-Oncogénicas c-crk/metabolismo , Pirazoles/química
14.
J Biomol Screen ; 16(1): 53-64, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20938045

RESUMEN

A 2-step kinase assay was developed and used in a high-throughput screen (HTS) of more than 1 million compounds in an effort to identify c-Abl tyrosine kinase activators. This assay employed a 2-step phosphorylation reaction: in the first step, purified recombinant c-Abl was activated by incubating with compound in the presence of adenosine triphosphate (ATP). In the second step, the TAMRA-labeled IMAP Abltide substrate was added to allow phosphorylation of the substrate to occur. The assay was calibrated such that inactive c-Abl protein was activated by ATP alone to a degree that it not only demonstrated a measurable c-Abl activity but also maintained a robust assay window for screening. The screen resulted in 8624 primary hits with >30% response. Further analysis showed that 1024 had EC(50) <10 µM with a max % response of >50%. These hits were structurally and chemically diverse with possibly different mechanisms for activating c-Abl. In addition, selective hits were shown to be cell permeable and were able to induce c-Abl activation as determined by In-Cell Western (ICW) analysis of HEK-MSRII cells transduced with BacMam virus expressing full-length c-Abl.


Asunto(s)
Activadores de Enzimas/farmacología , Ensayos Analíticos de Alto Rendimiento/métodos , Proteínas Proto-Oncogénicas c-abl/agonistas , Proteínas Proto-Oncogénicas c-abl/metabolismo , Adenosina Trifosfato/metabolismo , Baculoviridae/genética , Bioensayo , Descubrimiento de Drogas , Vectores Genéticos/genética , Células HEK293 , Humanos , Fosforilación , Bibliotecas de Moléculas Pequeñas/farmacología , Transfección
15.
Bioorg Med Chem Lett ; 19(1): 27-30, 2009 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19058966

RESUMEN

Endothelial lipase (EL) activity has been implicated in HDL catabolism, vascular inflammation, and atherogenesis, and inhibitors are therefore expected to be useful for the treatment of cardiovascular disease. Sulfonylfuran urea 1 was identified in a high-throughput screening campaign as a potent and non-selective EL inhibitor. A lead optimization effort was undertaken to improve potency and selectivity, and modifications leading to improved LPL selectivity were identified. Radiolabeling studies were undertaken to establish the mechanism of action for these inhibitors, which were ultimately demonstrated to be irreversible inhibitors.


Asunto(s)
Furanos , Lipasa/antagonistas & inhibidores , Compuestos de Sulfonilurea/síntesis química , Animales , Enfermedades Cardiovasculares/tratamiento farmacológico , Descubrimiento de Drogas , Evaluación Preclínica de Medicamentos , Endotelio/enzimología , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Humanos , Compuestos de Sulfonilurea/farmacología
16.
Mol Biotechnol ; 39(2): 127-34, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18327553

RESUMEN

Cortisol is an important glucocorticoid in humans that regulates many physiological processes. Human 11beta-hydroxysteroid dehydrogenase type 1 (11beta-HSD1) converts cortisone to cortisol in vivo and has emerged as an appealing therapeutic target for treating metabolic diseases. Here, we report a sensitive and robust high-throughput (HT) cell-based assay for screening 11beta-HSD1 inhibitors. This assay utilizes a HEK293 cell line transduced by a BacMam virus expressing human 11beta-HSD1. The enzyme activity in the cells was measured by quantifying cortisol levels released into the cell culture supernatant via a competitive homogenous time-resolved fluorescence (HTRF) method. We show that 11beta-HSD1 activity in supernatant of BacMam-transduced HEK293 cells increases with 11beta-HSD1 BacMam virus load in a dose-dependent manner, and is comparable to the enzyme activity detected in differentiated mouse adipocytes. In addition, we show that co-expression of hexose-6-phosphate dehydrogenase (H6PDH) is not required for the enzyme to function effectively as an oxo-reductase. This assay has been developed in low-volume 384-well format and it is sensitive, robust, and amenable to HT screening.


Asunto(s)
Fluoroinmunoensayo/métodos , Riñón/enzimología , Transducción Genética/métodos , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/antagonistas & inhibidores , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/metabolismo , Células 3T3-L1 , Adipocitos/enzimología , Animales , Deshidrogenasas de Carbohidratos/metabolismo , Supervivencia Celular , Cortisona/metabolismo , Medios de Cultivo/análisis , Humanos , Hidrocortisona/metabolismo , Ratones
17.
Genes Dev ; 17(13): 1581-91, 2003 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-12815072

RESUMEN

The nuclear bile acid receptor FXR has been proposed to play a central role in the feedback repression of the gene encoding cholesterol 7 alpha-hydroxylase (CYP7A1), the first and rate-limiting step in the biosynthesis of bile acids. We demonstrate that FXR directly regulates expression of fibroblast growth factor-19 (FGF-19), a secreted growth factor that signals through the FGFR4 cell-surface receptor tyrosine kinase. In turn, FGF-19 strongly suppresses expression of CYP7A1 in primary cultures of human hepatocytes and mouse liver through a c-Jun N-terminal kinase (JNK)-dependent pathway. This signaling cascade defines a novel mechanism for feedback repression of bile acid biosynthesis and underscores the vital role of FXR in the regulation of multiple pathways of cholesterol catabolism in the liver.


Asunto(s)
Ácidos y Sales Biliares/biosíntesis , Colesterol 7-alfa-Hidroxilasa/genética , Proteínas de Unión al ADN/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Hepatocitos/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Animales , Antracenos/farmacología , Línea Celular , Células Cultivadas , Ácido Quenodesoxicólico/farmacología , Colesterol 7-alfa-Hidroxilasa/metabolismo , Proteínas de Unión al ADN/agonistas , Proteínas de Unión al ADN/genética , Represión Enzimática , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/farmacología , Regulación de la Expresión Génica , Humanos , Isoxazoles/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos , Ratones , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-jun/metabolismo , Receptores Citoplasmáticos y Nucleares , Proteínas Recombinantes/farmacología , Elementos de Respuesta , Factores de Transcripción/agonistas , Factores de Transcripción/genética , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...