Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
PLoS One ; 16(3): e0248298, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33705488

RESUMEN

BACKGROUND: The combination of JAK/STAT and HDAC inhibitors exerted beneficial effects in haematological malignancies, presenting promising therapeutic CTCL targets. We aim to investigate the efficacy of JAK1/2i ruxolitinib in combination with HDACi resminostat in CTCL in vitro. MATERIAL & METHODS: Non-toxic concentrations of ruxolitinib and/or resminostat were administered to MyLa (MF) and SeAx (SS) cells for 24h. Cytotoxicity, cell proliferation and apoptosis were estimated through MTT, BrdU/7AAD and Annexin V/PI assay. Multi-pathway analysis was performed to investigate the effect of JAK1/2i and/or HDACi on JAK/STAT, Akt/mTOR and MAPK signalling pathways. RESULTS: Both drugs and their combination were cytotoxic in MyLa (p<0.05) and in SeAx cell line (p<0.001), inhibited proliferation of MyLa (p<0.001) and SeAx (p<0.001) at 24h, compared to untreated cells. Moreover, combined drug treatment induced apoptosis after 24h (p<0.001) in MyLa, and SeAx (p<0.001). The combination of drugs had a strong synergistic effect with a CI<1. Importantly, the drugs' combination inhibited phosphorylation of STAT3 (p<0.001), Akt (p<0.05), ERK1/2 (p<0.001) and JNK (p<0.001) in MyLa, while it reduced activation of Akt (p<0.05) and JNK (p<0.001) in SeAx. CONCLUSION: The JAKi/HDACi combination exhibited substantial anti-tumor effects in CTCL cell lines, and may represent a promising novel therapeutic modality for CTCL patients.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Linfoma Cutáneo de Células T/tratamiento farmacológico , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteínas de Neoplasias/metabolismo , Línea Celular Tumoral , Sinergismo Farmacológico , Humanos , Ácidos Hidroxámicos/agonistas , Ácidos Hidroxámicos/farmacología , Linfoma Cutáneo de Células T/metabolismo , Linfoma Cutáneo de Células T/patología , Nitrilos , Pirazoles/agonistas , Pirazoles/farmacología , Pirimidinas , Sulfonamidas/agonistas , Sulfonamidas/farmacología
2.
Microb Pathog ; 144: 104204, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32315753

RESUMEN

Toxoplasmosis is an infectious disease caused by the intracellular parasite Toxoplasma gondii that harms the brain and increases the risk of epilepsy acquisition. It is well known that cannabinoid (CB) signaling is activated following brain insults and protects the neurons from excitotoxicity and inflammation. We examined the role of CB neurotransmission in the proconvulsant effect of Toxoplasmosis in mice. Toxoplasmosis was established in mice by intraperitoneal injection of T. gondii cysts. The mice with acute and/or chronic Toxoplasma infection were pretreated (through intracerebroventricular injection) with CB1 and CB2 receptor agonists (ACEA and HU308) and antagonists (AM251 and AM630), as well as JZL184 (the irreversible inhibitor of mono acyl glycerol lipase, enzyme degrading the endogenous cannabinoid 2-Acyl glycerol). The seizure threshold was then measured by tail vein infusion of pentylenetetrazole. In healthy uninfected mice JZL184, ACEA, and AM630 increased the seizure threshold in a dose-dependent manner, whereas AM251 and HU308 showed dose-dependent proconvulsant effect. Mice with acute and/or chronic infection had a substantial lower seizure threshold than the uninfected mice. JZL 184, ACEA and AM630 inhibited proconvulsant effect of Toxoplasmosis, while AM251 and HU308 intensified proconvulsant effect of Toxoplasmosis. CB receptors play a role in proconvulsant effect of Toxoplasmosis in mice.


Asunto(s)
Receptores de Cannabinoides/efectos de los fármacos , Receptores de Cannabinoides/metabolismo , Toxoplasmosis/metabolismo , Animales , Benzodioxoles , Cannabinoides , Modelos Animales de Enfermedad , Indoles/agonistas , Masculino , Ratones , Piperidinas/agonistas , Pirazoles/agonistas , Receptor Cannabinoide CB1/efectos de los fármacos , Receptor Cannabinoide CB2/efectos de los fármacos , Toxoplasma
3.
Br J Haematol ; 191(1): 62-76, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32314355

RESUMEN

Multiple myeloma stem-like cells (MMSCs) are responsible for initiation and relapse, though novel treatment paradigms that effectively eradicate MMSCs are yet to be developed. Selective inhibition of the cell cycle regulatory kinase Wee1 by MK1775 is being explored as a potential anti-cancer therapeutic. We report that higher expression of Wee1 is correlated with poor survival in multiple myeloma (MM). The MM models and patient-derived CD138+ plasma cells are particularly sensitive to the growth-inhibitory effects of the Wee1 inhibitor MK1775. MK1775 induces Mus81-Eme1 endonuclease-mediated DNA damage in S-phase cell cycle that results in a blockade of replication and then apoptosis. Furthermore, MK1775 strongly suppresses the features of stemness in vitro, in vivo and in primary CD138+ cells by decreasing ALDH1+ cell fraction and the expression of ALDH1. In addition, co-treatment of MK1775 with bortezomib is synergistic in vitro and in vivo. Bortezomib, although it enhances ALDH1+ cells, when combined with MK1775 abrogates this stimulatory effect on stemness. Considering MM as an invariably incurable malignancy due to the presence of heterogenic myeloma stem-like cells, our study presents inhibition of Wee1 as a promising targeted therapy for MM and provides a compelling rationale to further investigate the activity of MK1775 against myeloma in clinical settings.


Asunto(s)
Bortezomib/farmacología , Proteínas de Ciclo Celular/antagonistas & inhibidores , Mieloma Múltiple , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Pirazoles/farmacología , Pirimidinonas/farmacología , Animales , Bortezomib/agonistas , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Sinergismo Farmacológico , Humanos , Ratones , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/enzimología , Proteínas de Neoplasias/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Pirazoles/agonistas , Pirimidinonas/agonistas , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Haematologica ; 102(9): 1587-1593, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28619845

RESUMEN

Fluorizoline is a new synthetic molecule that induces apoptosis by selectively targeting prohibitins. In the study herein, the pro-apoptotic effect of fluorizoline was assessed in 34 primary samples from patients with chronic lymphocytic leukemia. Fluorizoline induced apoptosis in chronic lymphocytic leukemia cells at concentrations in the low micromolar range. All primary samples were sensitive to fluorizoline irrespective of patients' clinical or genetic features, whereas normal T lymphocytes were less sensitive. Fluorizoline increased the protein levels of the pro-apoptotic B-cell lymphoma 2 family member NOXA in chronic lymphocytic leukemia cells. Furthermore, fluorizoline synergized with ibrutinib, 5-aminoimidazole-4-carboxamide riboside or venetoclax to induce apoptosis. These results suggest that targeting prohibitins could be a new therapeutic strategy for chronic lymphocytic leukemia.


Asunto(s)
Aminoimidazol Carboxamida/análogos & derivados , Apoptosis/efectos de los fármacos , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Hidrocarburos Fluorados/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Pirazoles/farmacología , Pirimidinas/farmacología , Proteínas Represoras/metabolismo , Ribonucleósidos/farmacología , Sulfonamidas/farmacología , Tiazolidinas/farmacología , Regulación hacia Arriba/efectos de los fármacos , Adenina/análogos & derivados , Aminoimidazol Carboxamida/agonistas , Aminoimidazol Carboxamida/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/agonistas , Sinergismo Farmacológico , Femenino , Humanos , Hidrocarburos Fluorados/agonistas , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Leucemia Linfocítica Crónica de Células B/metabolismo , Leucemia Linfocítica Crónica de Células B/patología , Masculino , Piperidinas , Prohibitinas , Pirazoles/agonistas , Pirimidinas/agonistas , Ribonucleósidos/agonistas , Sulfonamidas/agonistas , Tiazolidinas/agonistas , Células Tumorales Cultivadas
5.
Biochem Pharmacol ; 107: 91-100, 2016 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-26970402

RESUMEN

The human aryl hydrocarbon receptor is a cytosolic signaling molecule which affects immune response and aberrant cell growth. Canonical signaling of the receptor requires the recruitment of coactivators to the promoter region to remodel local chromatin structure. We predicted that interference of this recruitment would block the aryl hydrocarbon receptor function. To prove that, we employed phage display to identify nine peptides of twelve-amino-acid in length which target the C-terminal half of the human aryl hydrocarbon receptor, including the region where coactivators bind. Eight 12mer peptides, in the form of GFP fusion, suppressed the ligand-dependent transcription of six AHR target genes (cyp1a1, cyp1a2, cyp1b1, ugt1a1, nqo1, and ahrr) in different patterns in Hep3B cells, whereas the AHR antagonist CH-223191 suppressed all these target genes similarly. Three of the 12mer peptides (namely 11-3, 1-7, and 7-3) suppressed the 3MC-induced, CYP1A1-dependent EROD activity and the ROS production caused by benzo[a]pyrene. These 12mer peptides suppressed the AHR function synergistically with CH-223191. In conclusion, we provide evidence that targeting the C-terminal half of the human aryl hydrocarbon receptor is a viable, new approach to selectively block the receptor function.


Asunto(s)
Antineoplásicos/farmacología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/antagonistas & inhibidores , Carcinoma Hepatocelular/tratamiento farmacológico , Diseño de Fármacos , Hepatocitos/efectos de los fármacos , Proteínas de Neoplasias/antagonistas & inhibidores , Oligopéptidos/farmacología , Receptores de Hidrocarburo de Aril/antagonistas & inhibidores , Antineoplásicos/química , Antineoplásicos/metabolismo , Compuestos Azo/agonistas , Compuestos Azo/farmacología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/química , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Unión Competitiva , Carcinógenos/antagonistas & inhibidores , Carcinógenos/toxicidad , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Ligandos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Oligopéptidos/genética , Oligopéptidos/metabolismo , Fragmentos de Péptidos/antagonistas & inhibidores , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Biblioteca de Péptidos , Dominios y Motivos de Interacción de Proteínas , Pirazoles/agonistas , Pirazoles/farmacología , Especies Reactivas de Oxígeno/metabolismo , Receptores de Hidrocarburo de Aril/química , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/metabolismo , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/farmacología
7.
Leuk Res ; 39(1): 65-71, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25465126

RESUMEN

Interactions between the dual BCR/ABL and Src inhibitor bosutinib and the Chk1 inhibitor PF-00477736 were examined in BCR/ABL(+) leukemia cells, particularly imatinib-resistant cells, including those with the T315I mutation. Bosutinib blocked PF-00477736-induced ERK1/2 activation and sharply increased apoptosis in association with Mcl-1 inhibition, p34(cdc2) dephosphorylation, BimEL up-regulation, and DNA damage in imatinib-resistant CML or Ph(+) ALL cell lines. Inhibition of Src or MEK1 by shRNA significantly enhanced PF-0047736 lethality. Bosutinib/PF-00477736 co-treatment also potentiated cell death in CD34(+) CML patient samples, including dasatinib-resistant blast crisis cells exhibiting both T315I and E355G mutations, but was minimally toxic to normal CD34(+) cells. Finally, combined in vivo treatment significantly suppressed BaF3/T315I tumor growth and prolonged survival in an allogeneic mouse model. Together, these findings suggest that this targeted combination strategy warrants attention in IM-resistant CML or Ph(+) ALL.


Asunto(s)
Compuestos de Anilina , Antineoplásicos/farmacología , Benzamidas , Benzodiazepinonas , Resistencia a Antineoplásicos/efectos de los fármacos , Proteínas de Fusión bcr-abl/metabolismo , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Nitrilos , Piperazinas , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Inhibidores de Proteínas Quinasas , Proteínas Quinasas/metabolismo , Pirazoles , Pirimidinas , Quinolinas , Sustitución de Aminoácidos , Compuestos de Anilina/agonistas , Compuestos de Anilina/farmacología , Animales , Benzodiazepinonas/agonistas , Benzodiazepinonas/farmacología , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Resistencia a Antineoplásicos/genética , Sinergismo Farmacológico , Proteínas de Fusión bcr-abl/antagonistas & inhibidores , Proteínas de Fusión bcr-abl/genética , Humanos , Mesilato de Imatinib , Células K562 , Leucemia Linfocítica Crónica de Células B/genética , Leucemia Linfocítica Crónica de Células B/metabolismo , Leucemia Linfocítica Crónica de Células B/patología , MAP Quinasa Quinasa 1/genética , MAP Quinasa Quinasa 1/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/genética , Ratones , Ratones Desnudos , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Mutación Missense , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Nitrilos/agonistas , Nitrilos/farmacología , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Pirazoles/agonistas , Pirazoles/farmacología , Quinolinas/agonistas , Quinolinas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Acta Neuropathol ; 125(6): 795-813, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23604588

RESUMEN

In neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD) and prion diseases, deposits of aggregated disease-specific proteins are found. Oligomeric aggregates are presumed to be the key neurotoxic agent. Here we describe the novel oligomer modulator anle138b [3-(1,3-benzodioxol-5-yl)-5-(3-bromophenyl)-1H-pyrazole], an aggregation inhibitor we developed based on a systematic high-throughput screening campaign combined with medicinal chemistry optimization. In vitro, anle138b blocked the formation of pathological aggregates of prion protein (PrP(Sc)) and of α-synuclein (α-syn), which is deposited in PD and other synucleinopathies such as dementia with Lewy bodies (DLB) and multiple system atrophy (MSA). Notably, anle138b strongly inhibited all prion strains tested including BSE-derived and human prions. Anle138b showed structure-dependent binding to pathological aggregates and strongly inhibited formation of pathological oligomers in vitro and in vivo both for prion protein and α-synuclein. Both in mouse models of prion disease and in three different PD mouse models, anle138b strongly inhibited oligomer accumulation, neuronal degeneration, and disease progression in vivo. Anle138b had no detectable toxicity at therapeutic doses and an excellent oral bioavailability and blood-brain-barrier penetration. Our findings indicate that oligomer modulators provide a new approach for disease-modifying therapy in these diseases, for which only symptomatic treatment is available so far. Moreover, our findings suggest that pathological oligomers in neurodegenerative diseases share structural features, although the main protein component is disease-specific, indicating that compounds such as anle138b that modulate oligomer formation by targeting structure-dependent epitopes can have a broad spectrum of activity in the treatment of different protein aggregation diseases.


Asunto(s)
Encéfalo/efectos de los fármacos , Enfermedad de Parkinson/terapia , Enfermedades por Prión/terapia , Priones/efectos de los fármacos , Pirazoles/agonistas , Pirimidinas/agonistas , Animales , Encéfalo/metabolismo , Encéfalo/patología , Técnicas de Cultivo de Célula , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/metabolismo , Enfermedades por Prión/etiología , Enfermedades por Prión/metabolismo , Priones/metabolismo , Rotenona/farmacología , alfa-Sinucleína/farmacología
10.
PLoS One ; 8(3): e57523, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23520471

RESUMEN

Sarcomas are rare and heterogeneous mesenchymal tumors affecting both pediatric and adult populations with more than 70 recognized histologies. Doxorubicin and ifosfamide have been the main course of therapy for treatment of sarcomas; however, the response rate to these therapies is about 10-20% in metastatic setting. Toxicity with the drug combination is high, response rates remain low, and improvement in overall survival, especially in the metastatic disease, remains negligible and new agents are needed. Wee1 is a critical component of the G2/M cell cycle checkpoint control and mediates cell cycle arrest by regulating the phosphorylation of CDC2. Inhibition of Wee1 by MK1775 has been reported to enhance the cytotoxic effect of DNA damaging agents in different types of carcinomas. In this study we investigated the therapeutic efficacy of MK1775 in various sarcoma cell lines, patient-derived tumor explants ex vivo and in vivo both alone and in combination with gemcitabine, which is frequently used in the treatment of sarcomas. Our data demonstrate that MK1775 treatment as a single agent at clinically relevant concentrations leads to unscheduled entry into mitosis and initiation of apoptotic cell death in all sarcomas tested. Additionally, MK1775 significantly enhances the cytotoxic effect of gemcitabine in sarcoma cells lines with different p53 mutational status. In patient-derived bone and soft tissue sarcoma samples we showed that MK1775 alone and in combination with gemcitabine causes significant apoptotic cell death. Magnetic resonance imaging (MRI) and histopathologic studies showed that MK1775 induces significant cell death and terminal differentiation in a patient-derived xenograft mouse model of osteosarcoma in vivo. Our results together with the high safety profile of MK1775 strongly suggest that this drug can be used as a potential therapeutic agent in the treatment of both adult as well as pediatric sarcoma patients.


Asunto(s)
Antimetabolitos Antineoplásicos , Proteínas de Ciclo Celular/antagonistas & inhibidores , Desoxicitidina/análogos & derivados , Neoplasias Femorales/tratamiento farmacológico , Proteínas Nucleares/antagonistas & inhibidores , Osteosarcoma/tratamiento farmacológico , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Pirazoles , Pirimidinas , Adolescente , Adulto , Animales , Antimetabolitos Antineoplásicos/agonistas , Antimetabolitos Antineoplásicos/farmacología , Muerte Celular , Diferenciación Celular , Línea Celular Tumoral , Niño , Preescolar , Desoxicitidina/agonistas , Desoxicitidina/farmacología , Sinergismo Farmacológico , Femenino , Neoplasias Femorales/patología , Humanos , Masculino , Ratones , Ratones SCID , Persona de Mediana Edad , Trasplante de Neoplasias , Osteosarcoma/patología , Pirazoles/agonistas , Pirazoles/farmacología , Pirimidinas/agonistas , Pirimidinas/farmacología , Pirimidinonas , Trasplante Heterólogo , Ensayos Antitumor por Modelo de Xenoinjerto , Gemcitabina
11.
Life Sci ; 86(13-14): 510-7, 2010 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-20153751

RESUMEN

AIMS: Combinations of non-steroidal anti-inflammatory drugs (NSAIDs) and cannabinoids are promising because of their potential synergistic effects in analgesia, resulting in a reduction in dosage and minimizing adverse reactions. The analgesic effect of acetylsalicylic acid (ASA), probably due to a central mechanism, also implicates changes in the central monoaminergic system. Therefore, we decided to evaluate the antinociceptive interaction between the CB(1) receptor agonist, HU210, and ASA in tests involving central pain in rats as well as the implication of the central serotonergic system thereon. MAIN METHODS: The selective CB(1) antagonist SR141716A and the potent cannabinoid agonist HU210 were evaluated alone and in combination with ASA in both algesimetric tests (hot-plate and formalin tests) and for 5-HT activity and 5-HT(2) receptor density and affinity. KEY FINDINGS: ASA or HU210 alone showed a dose-dependent effect in both tests. HU210, at an inactive dose, significantly increased the antinociceptive effect of the sub-active dose of ASA. SR141716A (1.5mg/kgi.p.) was ineffective per se and failed to modify antinociception induced by the HU210 plus ASA combination in either test. HU210 plus ASA significantly decreased the 5-HIAA/5-HT ratio and the 5-HT(2) receptor density in the frontal cortex, changes not antagonized by SR141716A. SIGNIFICANCE: The present study provides evidence that mutual potentiation of the antinociceptive effects of HU210 and ASA may, at least partly, depend on serotonergic mechanisms, with an indirect participation of cannabinodiergic mechanism. In conclusion, combinations of low doses of cannabinoids and NSAIDs may be of interest from the therapeutic point of view.


Asunto(s)
Analgésicos/farmacología , Aspirina/farmacología , Lóbulo Frontal/efectos de los fármacos , Receptor Cannabinoide CB1 , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Animales , Cromatografía Líquida de Alta Presión , Masculino , Piperidinas/agonistas , Piperidinas/antagonistas & inhibidores , Pirazoles/agonistas , Pirazoles/antagonistas & inhibidores , Ratas , Receptor Cannabinoide CB1/agonistas , Receptor Cannabinoide CB1/antagonistas & inhibidores , Rimonabant
12.
Mol Imaging Biol ; 11(6): 386-98, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19330384

RESUMEN

BACKGROUND: Activation of microglia cells plays an important role in neurological diseases. Positron emission tomography (PET) with [(11)C]-(R)-PK11195 has already been used to visualize activated microglia cells in neurological diseases. However, [(11)C]-(R)-PK11195 may not possess the required sensitivity to visualize mild neuroinflammation. In this study, we evaluated the PET tracers [(11)C]-DPA-713 and [(18)F]-DPA-714 as agents for imaging of activated microglia in a rat model of herpes encephalitis. MATERIALS AND METHODS: Rats were intranasally inoculated with HSV-1. On day 6 or 7 after inoculation, small animal PET studies were performed to compare [(11)C]-(R)-PK11195, [(11)C]-DPA-713, and [(18)F]-DPA-714. RESULTS: Uptake of [(11)C]-DPA-713 in infected brain areas was comparable to that of [(11)C]-(R)-PK11195, but [(11)C]-DPA-713 showed lower non-specific binding. Non-specific uptake of [(18)F]-DPA-714 was lower than that of [(11)C]-(R)-PK11195. In the infected brain, total [(18)F]-DPA-714 uptake was lower than that of [(11)C]-(R)-PK11195, with comparable specific uptake. CONCLUSIONS: [(11)C]-DPA-713 may be more suitable for visualizing mild inflammation than [(11)C]-(R)-PK11195. In addition, the fact that [(18)F]-DPA-714 is an agonist PET tracer opens new possibilities to evaluate different aspects of neuroinflammation. Therefore, both tracers warrant further investigation in animal models and in a clinical setting.


Asunto(s)
Acetamidas/metabolismo , Proteínas Portadoras/metabolismo , Encefalitis por Herpes Simple/diagnóstico por imagen , Isoquinolinas/metabolismo , Pirazoles/agonistas , Pirazoles/metabolismo , Pirimidinas/agonistas , Pirimidinas/metabolismo , Receptores de GABA-A/metabolismo , Acetamidas/química , Animales , Animales no Consanguíneos , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Radioisótopos de Carbono/metabolismo , Modelos Animales de Enfermedad , Encefalitis por Herpes Simple/metabolismo , Radioisótopos de Flúor/metabolismo , Inmunohistoquímica , Isoquinolinas/química , Masculino , Estructura Molecular , Tomografía de Emisión de Positrones/métodos , Pirazoles/química , Pirimidinas/química , Ensayo de Unión Radioligante , Radiofármacos/metabolismo , Ratas , Ratas Wistar
14.
J Am Coll Cardiol ; 46(11): 2069-75, 2005 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-16325044

RESUMEN

OBJECTIVES: Regadenoson, a selective A2A adenosine receptor agonist, was evaluated for tolerability and effectiveness as a pharmacological stress agent for detecting reversible myocardial hypoperfusion when combined with single-photon emission computed tomography (SPECT). BACKGROUND: Adenosine and dipyridamole are nonselective adenosine agonists currently used as pharmacologic stressors. Despite proven safety, these agents often cause undesirable side effects and require a continuous infusion. METHODS: This Phase II, multicenter, open-label trial was conducted in 36 patients who had demonstrated ischemia on a 6-min adenosine SPECT imaging study within the previous 2 to 46 days. Patients received regadenoson as a rapid intravenous bolus dose of 400 microg (n = 18) or 500 microg (n = 18). The radiopharmaceutical was then delivered within one minute. The SPECT images were acquired in a standard manner and uniformly processed at a central laboratory. Regadenoson and adenosine studies were presented in random order and interpreted blindly with a 17-segment model by three observers. Additionally, quantitative analysis was performed with 4D-MSPECT software (University of Michigan, Ann Arbor, Michigan). RESULTS: Overall agreement for the presence of reversible hypoperfusion was 86%. The 400-mug dose was better tolerated. Overall, regadenoson was well-tolerated; side effects (e.g., chest discomfort, flushing, dyspnea) were generally mild in severity and self-limiting. High-grade atrioventricular block and bronchospasm were not observed. CONCLUSIONS: Regadenoson is well-tolerated and seems as effective as adenosine for detecting and quantifying the extent of hypoperfusion observed with SPECT perfusion imaging. Phase III clinical trials are now underway, given the promise of regadenoson's reduced side effects and simplicity of bolus administration.


Asunto(s)
Agonistas del Receptor de Adenosina A2 , Corazón/diagnóstico por imagen , Isquemia Miocárdica/diagnóstico por imagen , Tomografía Computarizada de Emisión de Fotón Único/métodos , Adenosina , Electrocardiografía , Humanos , Compuestos Organofosforados , Compuestos de Organotecnecio , Proyectos Piloto , Purinas/agonistas , Pirazoles/agonistas , Radiofármacos , Tecnecio Tc 99m Sestamibi
15.
Arch Environ Contam Toxicol ; 41(3): 325-32, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11503069

RESUMEN

Fipronil is a phenylpyrazole insecticide that is the active ingredient in the pesticide Icon 6.2 FS which is applied to rice seeds targeting the rice water weevil. An arthropod-selective insecticide, fipronil blocks the GABA-gated chloride channel and is unique in that several of its degradation products have been indicated to be equal or more potent than fipronil. After application of rice seeds (2-3 days postplant) to flooded rice fields, water is typically pumped from the rice fields and can be used for the culture of crayfish (Procambarus sp.). Because fipronil is selective for arthropods, is transported via organic sediment, and crayfish consume organic sediment, 96-h LC(50) experiments were conducted with fipronil and three of its environmental derivatives in crayfish under conditions without carrier solvents in water of similar pH, alkalinity, and hardness as observed in south Louisiana crayfish culture ponds. Measured LC(50)s for fipronil to red swamp (Procambarus clarkii) and white river (Procambarus zonangulus) crayfish were 14.3 (95% CI; 5.1-23.4) and 19.5 (95% CI; 11.1-27.9) microg/L, respectively. LC(50)s of fipronil sulfone (11.2; 9.2-13.2 microg/L), fipronil sulfide (15.5; 13-18 microg/L); and the photoproduct, desulfinyl fipronil (68.6; 46-95.2 microg/L) displayed very high toxicity in crayfish. In situ toxicity studies using caged crayfish in culturing ponds receiving effluent from drained rice fields indicated that effluent from rice fields planted with Icon-treated seed was significantly more toxic compared to untreated surface water (40% survival compared to 83% survival). Hazard quotient comparisons using measured water concentrations in the field and laboratory-based LC(50)s indicated that fipronil and its metabolites in water resulting from Icon-treated rice seed planting poses a significant risk to crayfish survival.


Asunto(s)
Astacoidea/efectos de los fármacos , Insecticidas/toxicidad , Pirazoles/toxicidad , Contaminantes Químicos del Agua/toxicidad , Animales , Astacoidea/fisiología , Exposición a Riesgos Ambientales , Oryza , Dinámica Poblacional , Pirazoles/agonistas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA