Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Cell Death Dis ; 15(5): 311, 2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38697987

RESUMEN

Cancer cells are highly dependent on bioenergetic processes to support their growth and survival. Disruption of metabolic pathways, particularly by targeting the mitochondrial electron transport chain complexes (ETC-I to V) has become an attractive therapeutic strategy. As a result, the search for clinically effective new respiratory chain inhibitors with minimized adverse effects is a major goal. Here, we characterize a new OXPHOS inhibitor compound called MS-L6, which behaves as an inhibitor of ETC-I, combining inhibition of NADH oxidation and uncoupling effect. MS-L6 is effective on both intact and sub-mitochondrial particles, indicating that its efficacy does not depend on its accumulation within the mitochondria. MS-L6 reduces ATP synthesis and induces a metabolic shift with increased glucose consumption and lactate production in cancer cell lines. MS-L6 either dose-dependently inhibits cell proliferation or induces cell death in a variety of cancer cell lines, including B-cell and T-cell lymphomas as well as pediatric sarcoma. Ectopic expression of Saccharomyces cerevisiae NADH dehydrogenase (NDI-1) partially restores the viability of B-lymphoma cells treated with MS-L6, demonstrating that the inhibition of NADH oxidation is functionally linked to its cytotoxic effect. Furthermore, MS-L6 administration induces robust inhibition of lymphoma tumor growth in two murine xenograft models without toxicity. Thus, our data present MS-L6 as an inhibitor of OXPHOS, with a dual mechanism of action on the respiratory chain and with potent antitumor properties in preclinical models, positioning it as the pioneering member of a promising drug class to be evaluated for cancer therapy. MS-L6 exerts dual mitochondrial effects: ETC-I inhibition and uncoupling of OXPHOS. In cancer cells, MS-L6 inhibited ETC-I at least 5 times more than in isolated rat hepatocytes. These mitochondrial effects lead to energy collapse in cancer cells, resulting in proliferation arrest and cell death. In contrast, hepatocytes which completely and rapidly inactivated this molecule, restored their energy status and survived exposure to MS-L6 without apparent toxicity.


Asunto(s)
Antineoplásicos , Proliferación Celular , Complejo I de Transporte de Electrón , Mitocondrias , Proteínas de Saccharomyces cerevisiae , Animales , Humanos , Complejo I de Transporte de Electrón/metabolismo , Complejo I de Transporte de Electrón/antagonistas & inhibidores , Antineoplásicos/farmacología , Ratones , Línea Celular Tumoral , Mitocondrias/metabolismo , Mitocondrias/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Desacopladores/farmacología , Fosforilación Oxidativa/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/efectos de los fármacos , Ratas , NADH Deshidrogenasa/metabolismo , NADH Deshidrogenasa/antagonistas & inhibidores
2.
Molecules ; 28(3)2023 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-36770826

RESUMEN

The chemokine receptor CXCR4 and its ligand CXCL12 regulate leukocyte trafficking, homeostasis and functions and are potential therapeutic targets in many diseases such as HIV-1 infection and cancers. Here, we identified new CXCR4 ligands in the CERMN chemical library using a FRET-based high-throughput screening assay. These are bis-imidazoline compounds comprising two imidazole rings linked by an alkyl chain. The molecules displace CXCL12 binding with submicromolar potencies, similarly to AMD3100, the only marketed CXCR4 ligand. They also inhibit anti-CXCR4 mAb 12G5 binding, CXCL12-mediated chemotaxis and HIV-1 infection. Further studies with newly synthesized derivatives pointed out to a role of alkyl chain length on the bis-imidazoline properties, with molecules with an even number of carbons equal to 8, 10 or 12 being the most potent. Interestingly, these differ in the functions of CXCR4 that they influence. Site-directed mutagenesis and molecular docking predict that the alkyl chain folds in such a way that the two imidazole groups become lodged in the transmembrane binding cavity of CXCR4. Results also suggest that the alkyl chain length influences how the imidazole rings positions in the cavity. These results may provide a basis for the design of new CXCR4 antagonists targeting specific functions of the receptor.


Asunto(s)
Imidazolinas , Transducción de Señal , Ligandos , Simulación del Acoplamiento Molecular , Receptores CXCR4 , Imidazoles/farmacología
3.
Microbiol Spectr ; 9(2): e0027421, 2021 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-34724729

RESUMEN

Human malaria infection begins with a one-time asymptomatic liver stage followed by a cyclic symptomatic blood stage. For decades, the research for novel antimalarials focused on the high-throughput screening of molecules that only targeted the asexual blood stages. In a search for new effective compounds presenting a triple action against erythrocytic and liver stages in addition to the ability to block the transmission of the disease via the mosquito vector, 2-amino-thienopyrimidinone derivatives were synthesized and tested for their antimalarial activity. One molecule, named gamhepathiopine (denoted as "M1" herein), was active at submicromolar concentrations against both erythrocytic (50% effective concentration [EC50] = 0.045 µM) and liver (EC50 = 0.45 µM) forms of Plasmodium falciparum. Furthermore, gamhepathiopine efficiently blocked the development of the sporogonic cycle in the mosquito vector by inhibiting the exflagellation step. Moreover, M1 was active against artemisinin-resistant forms (EC50 = 0.227 µM), especially at the quiescent stage. Nevertheless, in mice, M1 showed modest activity due to its rapid metabolization by P450 cytochromes into inactive derivatives, calling for the development of new parent compounds with improved metabolic stability and longer half-lives. These results highlight the thienopyrimidinone scaffold as a novel antiplasmodial chemotype of great interest to search for new drug candidates displaying multistage activity and an original mechanism of action with the potential to be used in combination therapies for malaria elimination in the context of artemisinin resistance. IMPORTANCE This work reports a new chemical structure that (i) displays activity against the human malaria parasite Plasmodium falciparum at 3 stages of the parasitic cycle (blood stage, hepatic stage, and sexual stages), (ii) remains active against parasites that are resistant to the first-line treatment recommended by the World Health Organization (WHO) for the treatment of severe malaria (artemisinins), and (iii) reduces transmission of the parasite to the mosquito vector in a mouse model. This new molecule family could open the way to the conception of novel antimalarial drugs with an original multistage mechanism of action to fight against Plasmodium drug resistance and block interhuman transmission of malaria.


Asunto(s)
Antimaláricos/farmacología , Malaria Falciparum/tratamiento farmacológico , Plasmodium cynomolgi/efectos de los fármacos , Plasmodium falciparum/efectos de los fármacos , Plasmodium yoelii/efectos de los fármacos , Pirimidinonas/farmacología , Animales , Antimaláricos/química , Artemisininas/farmacología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Perros , Resistencia a Medicamentos/fisiología , Femenino , Células Hep G2 , Humanos , Hígado/parasitología , Macaca fascicularis , Células de Riñón Canino Madin Darby , Masculino , Ratones , Ratones Endogámicos BALB C , Pirimidinonas/química
4.
Data Brief ; 33: 106492, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33294504

RESUMEN

Data presented in this article are associated with the research article "Identification of antiviral compounds against equid herpesvirus-1 using real-time cell assay screening: efficacy of decitabine and valganciclovir alone and in combination" [1]. These data correspond to the in vitro screening of 2,891 potential antiviral compounds against equid herpesvirus-1 (EHV-1) based on impedance measurements using the xCELLigence® RTCA MP System. This dataset includes compounds from three different libraries: i) 1,199 compounds from the Prestwick® Chemical Library, which contains mostly US Food and Drug Administration approved drugs (Prestwick® Chemical, Illkirch, France); ii) 1,651 compounds from the Centre d'Etudes et de Recherche sur le Médicament de Normandie (CERMN, Caen, France); iii) 41 compounds (called herein in-house antiviral library) selected for their effects against different human viruses. Compounds effective against EHV-1 were selected using the area under normalised curves (AUCn) and the time required for the Cell Index to decrease by 50% after virus infection (CIT50). The full dataset from the screen is made publicly available for further analyses.

5.
Cancers (Basel) ; 12(8)2020 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-32781579

RESUMEN

Paclitaxel is a microtubule stabilizing agent and a successful drug for cancer chemotherapy inducing, however, adverse effects. To reduce the effective dose of paclitaxel, we searched for pharmaceutics which could potentiate its therapeutic effect. We screened a chemical library and selected Carba1, a carbazole, which exerts synergistic cytotoxic effects on tumor cells grown in vitro, when co-administrated with a low dose of paclitaxel. Carba1 targets the colchicine binding-site of tubulin and is a microtubule-destabilizing agent. Catastrophe induction by Carba1 promotes paclitaxel binding to microtubule ends, providing a mechanistic explanation of the observed synergy. The synergistic effect of Carba1 with paclitaxel on tumor cell viability was also observed in vivo in xenografted mice. Thus, a new mechanism favoring paclitaxel binding to dynamic microtubules can be transposed to in vivo mouse cancer treatments, paving the way for new therapeutic strategies combining low doses of microtubule targeting agents with opposite mechanisms of action.

6.
Cell Death Differ ; 27(1): 117-129, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31068676

RESUMEN

Pro-survival stress-inducible chaperone HSP110 is the only HSP for which a mutation has been found in a cancer. Multicenter clinical studies demonstrated a direct association between HSP110 inactivating mutation presence and excellent prognosis in colorectal cancer patients. Here, we have combined crystallographic studies on human HSP110 and in silico modeling to identify HSP110 inhibitors that could be used in colorectal cancer therapy. Two molecules (foldamers 33 and 52), binding to the same cleft of HSP110 nucleotide-binding domain, were selected from a chemical library (by co-immunoprecipitation, AlphaScreening, Interference-Biolayer, Duo-link). These molecules block HSP110 chaperone anti-aggregation activity and HSP110 association to its client protein STAT3, thereby inhibiting STAT3 phosphorylation and colorectal cancer cell growth. These effects were strongly decreased in HSP110 knockdown cells. Foldamer's 33 ability to inhibit tumor growth was confirmed in two colorectal cancer animal models. Although tumor cell death (apoptosis) was noted after treatment of the animals with foldamer 33, no apparent toxicity was observed, notably in epithelial cells from intestinal crypts. Taken together, we identified the first HSP110 inhibitor, a possible drug-candidate for colorectal cancer patients whose unfavorable outcome is associated to HSP110.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/uso terapéutico , Neoplasias Colorrectales/tratamiento farmacológico , Proteínas del Choque Térmico HSP110/antagonistas & inhibidores , Animales , Antineoplásicos/toxicidad , Proliferación Celular , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Cristalografía por Rayos X , Proteínas del Choque Térmico HSP110/química , Proteínas del Choque Térmico HSP110/metabolismo , Humanos , Ratones , Modelos Moleculares , Factor de Transcripción STAT3/metabolismo
7.
Pestic Biochem Physiol ; 160: 11-19, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31519244

RESUMEN

The mite Varroa destructor is an ectoparasite and has been identified as a major cause of worldwide honey bee colony losses. The use of yearly treatments for the control of varroosis is the most common answer to prevent collapses of honey bee colonies due to the mite. However, the number of effective acaricides is small and the mite tends to become resistant to these few active molecules. In this study, we have been looking for a new original varroacide treatment inhibiting selectively Varroa destructor AChE (vdAChE) with respect to Apis mellifera AChE (amAChE). To do this an original drug design methodology was used applying virtual screening of the CERMN chemolibrary, starting from a vdAChE homology sequence model. By combining the in silico screening with in vitro experiments, two promising compounds were found. In vitro tests of AChE inhibition for both species have confirmed good selectivity toward the mite vdAChE. Moreover, an in vivo protocol was performed and highlighted a varroacide activity without acute consequences on honey bee survival. The two compounds discovered have the potential to become new drug leads for the development of new treatments against the mite varroa. The method described here clearly shows the potential of a drug-design approach to develop new solutions to safeguard honey bee health.


Asunto(s)
Acaricidas/farmacología , Varroidae/efectos de los fármacos , Acaricidas/química , Animales , Simulación por Computador , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología
8.
Eur J Med Chem ; 159: 357-380, 2018 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-30308410

RESUMEN

Protein-protein interactions are attractive targets because they control numerous cellular processes. In oncology, apoptosis regulating Bcl-2 family proteins are of particular interest. Apoptotic cell death is controlled via PPIs between the anti-apoptotic proteins hydrophobic groove and the pro-apoptotic proteins BH3 domain. In ovarian carcinoma, it has been previously demonstrated that Bcl-xL and Mcl-1 cooperate to protect tumor cells against apoptosis. Moreover, Mcl-1 is a key regulator of cancer cell survival and is a known resistance factor to Bcl-2/Bcl-xL pharmacological inhibitors making it an attractive therapeutic target. Here, using a structure-guided design from the oligopyridine lead Pyridoclax based on Noxa/Mcl-1 interaction we identified a new derivative, active at lower concentration as compared to Pyridoclax. This new derivative selectively binds to the Mcl-1 hydrophobic groove and releases Bak and Bim from Mcl-1 to induce cell death and sensitize cancer cells to Bcl-2/Bcl-xL targeting strategies.


Asunto(s)
Diseño de Fármacos , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Piridinas/farmacología , Muerte Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Simulación de Dinámica Molecular , Estructura Molecular , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/química , Unión Proteica , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-bcl-2/química , Piridinas/síntesis química , Piridinas/química , Relación Estructura-Actividad , Células Tumorales Cultivadas , Proteína bcl-X/antagonistas & inhibidores , Proteína bcl-X/metabolismo
9.
Antimicrob Agents Chemother ; 60(3): 1438-49, 2015 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-26666917

RESUMEN

In a search for new antifungal compounds, we screened a library of 4,454 chemicals for toxicity against the human fungal pathogen Aspergillus fumigatus. We identified sr7575, a molecule that inhibits growth of the evolutionary distant fungi A. fumigatus, Cryptococcus neoformans, Candida albicans, and Saccharomyces cerevisiae but lacks acute toxicity for mammalian cells. To gain insight into the mode of inhibition, sr7575 was screened against 4,885 S. cerevisiae mutants from the systematic collection of haploid deletion strains and 977 barcoded haploid DAmP (decreased abundance by mRNA perturbation) strains in which the function of essential genes was perturbed by the introduction of a drug resistance cassette downstream of the coding sequence region. Comparisons with previously published chemogenomic screens revealed that the set of mutants conferring sensitivity to sr7575 was strikingly narrow, affecting components of the endoplasmic reticulum-associated protein degradation (ERAD) stress response and the ER membrane protein complex (EMC). ERAD-deficient mutants were hypersensitive to sr7575 in both S. cerevisiae and A. fumigatus, indicating a conserved mechanism of growth inhibition between yeast and filamentous fungi. Although the unfolded protein response (UPR) is linked to ERAD regulation, sr7575 did not trigger the UPR in A. fumigatus and UPR mutants showed no enhanced sensitivity to the compound. The data from this chemogenomic analysis demonstrate that sr7575 exerts its antifungal activity by disrupting ER protein quality control in a manner that requires ERAD intervention but bypasses the need for the canonical UPR. ER protein quality control is thus a specific vulnerability of fungal organisms that might be exploited for antifungal drug development.


Asunto(s)
Antifúngicos/farmacología , Antifúngicos/toxicidad , Aspergillus fumigatus/efectos de los fármacos , Degradación Asociada con el Retículo Endoplásmico/efectos de los fármacos , Animales , Aspergillus fumigatus/genética , Candida albicans/efectos de los fármacos , Cryptococcus neoformans/efectos de los fármacos , Evaluación Preclínica de Medicamentos/métodos , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Degradación Asociada con el Retículo Endoplásmico/genética , Células HeLa/efectos de los fármacos , Humanos , Ratones Endogámicos , Pruebas de Sensibilidad Microbiana , Mutación , Saccharomyces cerevisiae/efectos de los fármacos , Saccharomyces cerevisiae/genética , Bibliotecas de Moléculas Pequeñas/farmacología , Respuesta de Proteína Desplegada/efectos de los fármacos
10.
Eur J Med Chem ; 95: 16-28, 2015 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-25791675

RESUMEN

A preliminary in vitro screening of compounds belonging to various chemical families from our library revealed the thieno[3,2-d]pyrimidin-4(3H)-one scaffold displayed a promising profile against Plasmodium falciparum. Then, 120 new derivatives were synthesized and evaluated in vitro; compared to drug references, 40 showed good activity toward chloroquine sensitive (IC50 35-344 nM) and resistant (IC50 45-800 nM) P. falciparum strains. They were neither cytotoxic (CC50 15-50 µM) toward HepG2 and CHO cells, nor mutagenic. Structure-activity relationships were defined. The lead-compound also appeared active against the Plasmodium liver stages (Plasmodium yoelii IC50 = 35 nM) and a preliminary in vivo evaluation indicated the in vitro activity was preserved (45% reduction in parasitemia compared to untreated infected mice). A mechanistic study demonstrated these molecules do not involve any of the pathways described for commercial drugs and exert a specific activity on the ring and trophozoite stages.


Asunto(s)
Antimaláricos/farmacología , Descubrimiento de Drogas , Eritrocitos/efectos de los fármacos , Hígado/efectos de los fármacos , Malaria/tratamiento farmacológico , Plasmodium falciparum/efectos de los fármacos , Pirimidinas/química , Animales , Antimaláricos/química , Células CHO , Proliferación Celular/efectos de los fármacos , Cricetinae , Cricetulus , Células Hep G2 , Humanos , Malaria/parasitología , Masculino , Ratones , Parasitemia/tratamiento farmacológico , Parasitemia/parasitología , Plasmodium falciparum/crecimiento & desarrollo , Relación Estructura-Actividad , Trofozoítos/efectos de los fármacos
11.
Chembiochem ; 16(3): 432-9, 2015 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-25619419

RESUMEN

Cyclin-dependent kinases (CDKs) control many cellular processes and are considered important therapeutic targets. Large collections of inhibitors targeting CDK active sites have been discovered, but their use in chemical biology or drug development has been often hampered by their general lack of specificity. An alternative approach to develop more specific inhibitors is targeting protein interactions involving CDKs. CKS proteins interact with some CDKs and play important roles in cell division. We discovered two small-molecule inhibitors of CDK-CKS interactions. They bind to CDK2, do not inhibit its enzymatic activity, inhibit the proliferation of tumor cell lines, induce an increase in G1 and/or S-phase cell populations, and cause a decrease in CDK2, cyclin A, and p27(Kip1) levels. These molecules should help decipher the complex contributions of CDK-CKS complexes in the regulation of cell division, and they might present an interesting therapeutic potential.


Asunto(s)
Quinasas CDC2-CDC28/metabolismo , Quinasa 2 Dependiente de la Ciclina/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología , Quinasas CDC2-CDC28/antagonistas & inhibidores , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular/metabolismo , División Celular/efectos de los fármacos , Ciclina A/antagonistas & inhibidores , Ciclina A/metabolismo , Quinasa 2 Dependiente de la Ciclina/metabolismo , Relación Dosis-Respuesta a Droga , Ensayos Analíticos de Alto Rendimiento , Humanos , Células MCF-7/efectos de los fármacos , Simulación del Acoplamiento Molecular , Estructura Molecular , Terapia Molecular Dirigida , Mapas de Interacción de Proteínas/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/metabolismo
12.
Eur J Med Chem ; 83: 26-35, 2014 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-24946216

RESUMEN

Thanks to a preliminary in vitro screening of several CCl3-substituted-nitrogen containing heterocycles belonging to our chemical library, the 2-trichloromethylquinoxaline scaffold appeared to be of potential interest for developing new antiplasmodial agents. Then, combining these experimental results to the antimalarial properties reported for various pyrrolo[1,2-a]quinoxaline derivatives, an original series of fifteen 7-substituted-4-trichoromethylpyrrolo[1,2-a]quinoxalines was synthesized in a 4 to 5 reaction steps pathway. All molecules were evaluated in vitro toward both their antiplasmodial activity on the K1 multi-resistant Plasmodium falciparum strain and their cytotoxicity on the HepG2 human cell line. Thus, 3 hit molecules were identified, displaying IC50 values in the micromolar range and low cytotoxicity values, reaching good selectivity indexes, in comparison with the reference drugs chloroquine and doxycycline. Structure-activity relationship studies showed that the pyrrolo[1,2-a]quinoxaline scaffold can support selective antiplasmodial activity when substituted at position 4 by a CCl3 group. However, substitution at position 7 of the same scaffold is neither beneficial for cytotoxicity nor favourable for the solubility in the biological media.


Asunto(s)
Antimaláricos/síntesis química , Antimaláricos/farmacología , Plasmodium falciparum/efectos de los fármacos , Quinoxalinas/síntesis química , Quinoxalinas/farmacología , Antimaláricos/química , Antimaláricos/toxicidad , Técnicas de Química Sintética , Células Hep G2 , Humanos , Metilación , Quinoxalinas/química , Quinoxalinas/toxicidad
13.
Mol Imaging ; 132014.
Artículo en Inglés | MEDLINE | ID: mdl-24622810

RESUMEN

The pharmacokinetic properties of radiotracers are crucial for successful in vivo single-photon emission computed tomographic (SPECT) imaging. Our goal was to determine if MDR1A-deficient animals could allow better SPECT imaging outcomes than wild-type (WT) animals for a selection of serotoninergic radioligands. Thus, we compared the performances of 123I-p-MPPI, 123I-R91150, 123I-SB207710, and 123I-ADAM radioligands, for imaging of their respective targets (5-hydroxytryptamine [5-HT]1A, 5-HT2A, 5-HT4, and serotonin transporter [SERT]), in WT and Mdr1a knockout (KO) rats. With 123I-SB207710, virtually no SPECT signal was recorded in the brain of WT or KO animals. For 123I-p-MPPI, low nondisplaceable binding potentials (BPND, mean ± SD) were observed in WT (0.49 ± 0.25) and KO (0.89 ± 0.52) animals. For 123I-ADAM, modest imaging contrast was observed in WT (1.27 ± 0.02) and KO (1.31 ± 0.09) animals. For 123I-R91150, the BPND were significantly higher in Mdr1a KO (3.98 ± 0.65) animals compared to WT animals (1.22 ± 0.26). The pharmacokinetics of 123I-SB207710 and 123I-p-MPPI do not make them ideal tracers for preclinical SPECT neuroimaging. 123I-ADAM showed adequate brain uptake regardless of Mdr1a expression and appeared suitable for preclinical SPECT neuroimaging in both animal strains. The use of Mdr1a KO animals significantly improved the brain penetration of 123I-R91150, making this animal strain an interesting option when considering SPECT neuroimaging of 5-HT2A receptors in rat.


Asunto(s)
Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Encéfalo/diagnóstico por imagen , Radioisótopos de Yodo/farmacocinética , Radiofármacos/farmacocinética , Receptores de Serotonina/metabolismo , Inhibidores Selectivos de la Recaptación de Serotonina/farmacocinética , Tomografía Computarizada de Emisión de Fotón Único/métodos , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Aminopiridinas/farmacocinética , Animales , Encéfalo/metabolismo , Cinanserina/análogos & derivados , Cinanserina/farmacocinética , Técnicas de Inactivación de Genes , Masculino , Especificidad de Órganos , Piperazinas/farmacocinética , Piperidinas/farmacocinética , Ratas , Ratas Sprague-Dawley , Distribución Tisular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA