Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros

Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Bioorg Med Chem ; 98: 117581, 2024 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-38176113

RESUMEN

Although KRAS protein had been classified as an undruggable target, inhibitors of KRAS G12C mutant protein were recently reported to show clinical efficacy in solid tumors. In our previous report, we identified 1-{2,7-diazaspiro[3.5]non-2-yl}prop-2-en-1-one derivative (1) as a KRAS G12C inhibitor that covalently binds to Cys12 of KRAS G12C protein. Compound 1 exhibited potent cellular pERK inhibition and cell growth inhibition against a KRAS G12C mutation-positive cell line and showed an antitumor effect on subcutaneous administration in an NCI-H1373 (KRAS G12C mutation-positive cell line) xenograft mouse model in a dose-dependent manner. In this report, we further optimized the substituents on the quinazoline scaffold based on the structure-based drug design from the co-crystal structure analysis of compound 1 and KRAS G12C to enhance in vitro activity. As a result, ASP6918 was found to exhibit extremely potent in vitro activity and induce dose-dependent tumor regression in an NCI-H1373 xenograft mouse model after oral administration.


Asunto(s)
Neoplasias Pulmonares , Neoplasias , Humanos , Animales , Ratones , Proteínas Proto-Oncogénicas p21(ras)/genética , Mutación , Relación Estructura-Actividad , Neoplasias Pulmonares/tratamiento farmacológico
2.
Br J Cancer ; 126(5): 744-753, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34795410

RESUMEN

BACKGROUND: KRAS is one of the most frequently mutated oncogenes in various cancers, and several novel KRAS G12C direct inhibitors are now in clinical trials. Here, we characterised the anti-tumour efficacy of ASP2453, a novel KRAS G12C inhibitor, in preclinical models of KRAS G12C-mutated cancer. METHODS: We evaluated the in vitro and in vivo activity of ASP2453, alone or in combination with targeted agents and immune checkpoint inhibitors, in KRAS G12C-mutated cancer cells and xenograft models. We also assessed pharmacological differences between ASP2453 and AMG 510, another KRAS G12C inhibitor, using an SPR assay, washout experiments and an AMG 510-resistant xenograft model. RESULTS: ASP2453 potently and selectively inhibited KRAS G12C-mediated growth, KRAS activation and downstream signalling in vitro and in vivo, and improved the anti-tumour effects of targeted agents and immune checkpoint inhibitors. Further, ASP2453 had more rapid binding kinetics to KRAS G12C protein and showed more potent inhibitory effects on KRAS activation and cell proliferation after washout than AMG 510. ASP2453 also induced tumour regression in an AMG 510-resistant xenograft model. CONCLUSIONS: ASP2453 is a potential therapeutic agent for KRAS G12C-mutated cancer. ASP2453 showed efficacy in AMG 510-resistant tumours, even among compounds with the same mode of action.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Resistencia a Antineoplásicos/efectos de los fármacos , Mutación , Piperazinas/administración & dosificación , Proteínas Proto-Oncogénicas p21(ras)/genética , Piridinas/administración & dosificación , Pirimidinas/administración & dosificación , Bibliotecas de Moléculas Pequeñas/administración & dosificación , Células A549 , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Línea Celular Tumoral , Células HCT116 , Humanos , Masculino , Ratones , Piperazinas/farmacología , Piridinas/farmacología , Pirimidinas/farmacología , Distribución Aleatoria , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Bioorg Med Chem ; 71: 116949, 2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-35926326

RESUMEN

RAS protein plays a key role in cellular proliferation and differentiation. RAS gene mutation is a known driver of oncogenic alternation in human cancer. RAS inhibition is an effective therapeutic treatment for solid tumors, but RAS protein has been classified as an undruggable target. Recent reports have demonstrated that a covalent binder to KRAS protein at a mutated cysteine residue (G12C) is effective for the treatment of solid tumors. Here, we report a series of 1-{2,7-diazaspiro[3.5]nonan-2-yl}prop-2-en-1-one derivatives as potent covalent inhibitors against KRAS G12C identified throughout structural optimization of an acryloyl amine moiety to improve in vitro inhibitory activity. From an X-ray complex structural analysis, the 1-{2,7-diazaspiro[3.5]nonan-2-yl}prop-2-en-1-one moiety binds in the switch-II pocket of KRAS G12C. Further optimization of the lead compound (5c) led to the successful identification of 1-[7-[6-chloro-8-fluoro-7-(5-methyl-1H-indazol-4-yl)-2-[(1-methylpiperidin-4-yl)amino]quinazolin-4-yl]-2,7-diazaspiro[3.5]nonan-2-yl]prop-2-en-1-one (7b), a potent compound with high metabolic stabilities in human and mouse liver microsomes. Compound 7b showed a dose-dependent antitumor effect on subcutaneous administration in an NCI-H1373 xenograft mouse model.


Asunto(s)
Alcanos/farmacología , Neoplasias , Proteínas Proto-Oncogénicas p21(ras) , Animales , Proliferación Celular , Humanos , Ratones , Mutación , Neoplasias/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/farmacología , Proteínas ras/genética , Proteínas ras/metabolismo
4.
Bioorg Med Chem ; 28(5): 115307, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-32007387

RESUMEN

Adenosine monophosphate (AMP)-activated protein kinase (AMPK) plays a key role in maintaining cellular metabolism. AMP or adenosine diphosphate (ADP) levels rise during metabolic stress, such as during nutrient starvation, hypoxia and muscle contraction, and bind to AMPK to induce activity. Recently, activation of AMPK has been considered an attractive therapeutic strategy in the field of human oncology. Structural optimization of lead compound 2, a new type of AMPK activator with potent AMPK activation activity and attractive selective growth inhibition against human cancer cells, improved aqueous solubility, metabolic stability and animal pharmacokinetics (PK) and culminated in the identification of (5-{1-[(6-methoxypyridin-3-yl)methyl]piperidin-4-yl}-1H-benzimidazol-2-yl)(4-{[4-(trifluoromethyl)phenyl]methyl}piperazin-1-yl)methanone ditosylate, ASP4132 (28). Studies on ASP4132 had advanced to clinical trials for the treatment of cancer.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Antineoplásicos/farmacología , Desarrollo de Medicamentos , Inhibidores de Proteínas Quinasas/farmacología , Administración Oral , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/química , 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 , Ratones , Ratones Desnudos , Estructura Molecular , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/química , Ratas , Ratas Sprague-Dawley , Relación Estructura-Actividad
5.
Chem Pharm Bull (Tokyo) ; 68(5): 452-465, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32378543

RESUMEN

This study reports the synthesis and evaluation of novel indirect AMP-activated protein kinase (AMPK) activators. The series of compounds selectively inhibited cell growth in several human breast cancer cell lines by activating AMPK. We performed back-up medicinal chemistry synthetic research on ASP4132, a previously reported as a compound for clinical development that acts as an indirect AMPK activator. This led to the successful identification of 4-({4-[5-({1-[(5-ethoxypyrazin-2-yl)methyl]-4-fluoropiperidin-4-yl}methoxy)-3-methylpyridine-2-carbonyl]piperazin-1-yl}methyl)benzonitrile succinate (27b), a potent, highly aqueous soluble and metabolically stable compound in human hepatocytes. Compound 27b also showed weaker human Ether-a-go-go Related Gene (hERG) inhibitory activity than that of compound 13 and ASP4132. Therefore, 27b was a promising AMPK activator and a second-generation clinical candidate for treatment for human cancer.


Asunto(s)
Antineoplásicos/farmacología , Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Canales de Potasio Éter-A-Go-Go/genética , Canales de Potasio Éter-A-Go-Go/metabolismo , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Masculino , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Desnudos , Microsomas Hepáticos/química , Microsomas Hepáticos/metabolismo , Estructura Molecular , Ratas , Ratas Sprague-Dawley , Solubilidad
6.
Chem Pharm Bull (Tokyo) ; 68(1): 77-90, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31902903

RESUMEN

Novel 3,5-dimethylpyridin-4(1H)-one scaffold compounds were synthesized and evaluated as AMP-activated protein kinase (AMPK) activators. Unlike direct AMPK activators, this series of compounds showed selective cell growth inhibitory activity against human breast cancer cell lines. By optimizing the lead compound (4a) from our library, 2-[({1'-[(4-fluorophenyl)methyl]-2-methyl-1',2',3',6'-tetrahydro[3,4'-bipyridin]-6-yl}oxy)methyl]-3,5-dimethylpyridin-4(1H)-one (25) was found to have potent AMPK activating activity. Compound 25 also showed good aqueous solubility while maintaining the unique selectivity in cell growth inhibitory activity.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Piridonas/química , Proteínas Quinasas Activadas por AMP/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Humanos , Piridonas/síntesis química , Piridonas/farmacología , Solubilidad , Relación Estructura-Actividad
7.
CPT Pharmacometrics Syst Pharmacol ; 10(8): 864-877, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34043291

RESUMEN

KRAS is a small GTPase family protein that relays extracellular growth signals to cell nucleus. KRASG12C mutations lead to constitutive proliferation signaling and are prevalent across human cancers. ASP2453 is a novel, highly potent, and selective inhibitor of KRASG12C . Although preclinical data suggested impressive efficacy, it remains unclear whether ASP2453 will show more favorable clinical response compared to more advanced competitors, such as AMG 510. Here, we developed a quantitative systems pharmacology (QSP) model linking KRAS signaling to tumor growth in patients with non-small cell lung cancer. The model was parameterized using in vitro ERK1/2 phosphorylation and in vivo xenograft data for ASP2453. Publicly disclosed clinical data for AMG 510 were used to generate a virtual population, and tumor size changes in response to ASP2453 and AMG 510 were simulated. The QSP model predicted ASP2453 exhibits greater clinical response than AMG 510, supporting potential differentiation and critical thinking for clinical trials.


Asunto(s)
Antineoplásicos , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Modelos Biológicos , Proteínas Proto-Oncogénicas p21(ras)/antagonistas & inhibidores , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/genética , Simulación por Computador , Humanos , Neoplasias Pulmonares/genética , Ratones , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Mutación , Farmacología en Red , Compuestos Orgánicos/administración & dosificación , Compuestos Orgánicos/farmacología , Fosforilación , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Mol Pharmacol ; 78(5): 961-70, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20736318

RESUMEN

Excessive hepatic glucose production through the gluconeogenesis pathway is partially responsible for the elevated glucose levels observed in patients with type 2 diabetes mellitus (T2DM). The forkhead transcription factor forkhead box O1 (Foxo1) plays a crucial role in mediating the effect of insulin on hepatic gluconeogenesis. Here, using a db/db mouse model, we demonstrate the effectiveness of Foxo1 inhibitor, an orally active small-molecule compound, as a therapeutic drug for treating T2DM. Using mass spectrometric affinity screening, we discovered a series of compounds that bind to Foxo1, identifying among them the compound, 5-amino-7-(cyclohexylamino)-1-ethyl-6-fluoro-4-oxo-1,4-dihydroquinoline-3-carboxylic acid (AS1842856), which potently inhibits human Foxo1 transactivation and reduces glucose production through the inhibition of glucose-6 phosphatase and phosphoenolpyruvate carboxykinase mRNA levels in a rat hepatic cell line. Oral administration of AS1842856 to diabetic db/db mice led to a drastic decrease in fasting plasma glucose level via the inhibition of hepatic gluconeogenic genes, whereas administration to normal mice had no effect on the fasting plasma glucose level. Treatment with AS1842856 also suppressed an increase in plasma glucose level caused by pyruvate injection in both normal and db/db mice. Taken together, these findings indicate that the Foxo1 inhibitor represents a new class of drugs for use in treating T2DM.


Asunto(s)
Factores de Transcripción Forkhead/antagonistas & inhibidores , Hiperglucemia/tratamiento farmacológico , Hipoglucemiantes/farmacología , Quinolonas/farmacología , Animales , Línea Celular Tumoral , Ayuno , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/genética , Glucosa/biosíntesis , Glucosa-6-Fosfatasa/antagonistas & inhibidores , Glucosa-6-Fosfatasa/genética , Humanos , Hiperglucemia/metabolismo , Hipoglucemiantes/uso terapéutico , Masculino , Espectrometría de Masas , Ratones , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Fosfoenolpiruvato Carboxiquinasa (GTP)/antagonistas & inhibidores , Fosfoenolpiruvato Carboxiquinasa (GTP)/genética , Ácido Pirúvico/farmacología , Quinolonas/uso terapéutico , ARN Mensajero/antagonistas & inhibidores , Ratas , Relación Estructura-Actividad , Activación Transcripcional
9.
Neuropharmacology ; 50(5): 558-67, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16388830

RESUMEN

Non-steroidal anti-inflammatory drugs (NSAIDs) are frequently used in the treatment of inflammation and pain. In many reports, NSAIDs have induced apoptosis in a variety of cell lines such as colon cancer cells. On the other hand, more recently a few reports have found that NSAIDs protect against apoptosis. Here we investigate endoplasmic reticulum (ER)-stress-induced apoptosis of neuronal cells. The aim of this study is to examine the involvement of NSAIDs, in particular diclofenac, on ER-stress-induced apoptosis of human neuroblastoma SH-SY5Y cells. Diclofenac significantly suppressed SH-SY5Y cell death induced by two types of ER-stress-inducing agents: thapsigargin, an inhibitor of Ca2+-ATPase on the endoplasmic reticulum membrane, and tunicamycin, a glycosylation blocker. Other NSAIDs, such as indomethacin, ibuprofen, aspirin, and ketoprofen, also suppressed ER-stress-induced SH-SY5Y cell death. The dose-dependent anti-apoptotic effect of diclofenac did not correlate with the reduction of prostaglandin release. Administration of prostaglandin E2, which was a primary product of arachidonic metabolism, showed no effects against anti-apoptotic effects produced by diclofenac. Thapsigargin and tunicamycin each significantly activated caspase-3, -9, and -2 in the intrinsic apoptotic pathway in SH-SY5Y cells. Diclofenac suppressed the activation of caspases induced by both ER stresses. Thapsigargin and tunicamycin decreased the mitochondrial membrane potential in SH-SY5Y cells. Diclofenac suppressed the mitochondrial depolarization induced by both ER stresses. Diclofenac inhibited ER-stress-induced apoptosis of SH-SY5Y cells by suppressing the activation of caspases in the intrinsic apoptotic pathway. This is the first report to find that diclofenac has protective effects against ER-stress-induced apoptosis.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Apoptosis/efectos de los fármacos , Caspasas/metabolismo , Diclofenaco/farmacología , Retículo Endoplásmico/fisiología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Fragmentación del ADN/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Retículo Endoplásmico/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Ensayo de Inmunoadsorción Enzimática/métodos , Técnica del Anticuerpo Fluorescente/métodos , Humanos , Neuroblastoma , Prostaglandinas/metabolismo , Tapsigargina/farmacología
10.
Neuropharmacology ; 50(8): 1041-7, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16574167

RESUMEN

Neurite outgrowth plays a key role in neuronal development and regeneration, and is the hallmark assay for the effects of neurotrophic factors such as nerve growth factor (NGF). However, measuring neurite outgrowth is a slow and resource-intensive process. We therefore wanted to identify surrogate biomarkers for neurite outgrowth activity by gene expression analysis in SH-O10 cells, a subclone of the human SH-SY5Y neuroblastoma cell line but with much higher NGF-induced neurite outgrowth activity. Microarray analysis identified seven genes where mRNA levels were changed. NGF-induced decreases in levels of two genes, CyclinB2 and BIRC5, were confirmed by quantitative real-time RT-PCR. Levels of NGF-induced decreases in CyclinB2 and BIRC5 mRNA in several SH-SY5Y subclones with different neurite outgrowth responses correlated with their neurite outgrowth activities. Decreases in CyclinB2 and BIRC5 mRNA induced by FK506 or retinoic acid, both of which exert potentiation of NGF-induced neurite outgrowth effects but with different mechanisms, also correlated with their neurite outgrowth activities. In conclusion, decreasing levels of CyclinB2 and BIRC5 mRNA strongly correlate with neurite outgrowth activities in terms of NGF-related effect in SH-SY5Y subclonal cells, and have potential to become quantitative surrogate biomarkers for measuring NGF-related neurite outgrowth.


Asunto(s)
Diferenciación Celular/genética , Ciclina B/genética , Proteínas Asociadas a Microtúbulos/genética , Proteínas de Neoplasias/genética , Neuritas/metabolismo , Neuroblastoma/patología , Antineoplásicos/farmacología , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Expresión Génica/efectos de los fármacos , Expresión Génica/fisiología , Humanos , Inmunosupresores/farmacología , Proteínas Inhibidoras de la Apoptosis , Modelos Lineales , Factor de Crecimiento Nervioso/farmacología , Neuritas/efectos de los fármacos , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Survivin , Tacrolimus/farmacología , Tretinoina/farmacología
11.
J Vet Med Sci ; 68(5): 507-10, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16757897

RESUMEN

Genes and proteins of human origin are often administered to monkeys for research purposes, however, it can be difficult to obtain sufficient levels of the products in vivo due to immunological clearance. In this study, we showed that human erythropoietin (hEPO) induces generation of anti-hEPO antibody in cynomolgus macaques (n=2), although 92% of amino acid residues are common between the human and macaque EPO. The administered hEPO was thus eliminated from the animals. On the other hand, when an immunosuppressant, cyclosporin A (CyA), was administered (6 mg/kg) intramuscularly every other day in combination with hEPO (n=2), no anti-hEPO antibody was generated and high serum levels of hEPO were obtained during administration of hEPO, resulting in an increase in serum hemoglobin levels. No adverse effects associated with CyA were observed. Thus, CyA treatment is useful for prevention of immune responses associated with the administration of human proteins in monkeys.


Asunto(s)
Anticuerpos/sangre , Ciclosporina/farmacología , Eritropoyetina/inmunología , Inmunosupresores/farmacología , Macaca fascicularis/inmunología , Animales , Formación de Anticuerpos/efectos de los fármacos , Femenino , Hemoglobinas/análisis , Humanos , Hipersensibilidad Inmediata/prevención & control , Hipersensibilidad Inmediata/veterinaria , Macaca fascicularis/sangre , Masculino
12.
Biochem Pharmacol ; 69(10): 1473-81, 2005 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-15857611

RESUMEN

Recently, we established an in vitro model of apoptosis induced by exposure of neuroblastoma SH-SY5Y cells to thapsigargin, an endoplasmic reticular calcium-ATPase inhibitor, and demonstrated that FK506 (tacrolimus) protected against apoptosis. The purpose of this paper was to investigate a possible correlation between the protective effect of FK506 against apoptosis and the regulation of the serum inducible kinase (SNK) and fibroblast growth factor inducible kinase (FNK) genes-which are polo-like kinases expressed abundantly in the brain by FK506. Thapsigargin increased the mRNA level of SNK and FNK in SH-SY5Y cells. FK506 inhibited the increase in SNK mRNA but not FNK mRNA. Deletion analysis of the SNK promoter showed that the promoter site, which was regulated by thapsigargin and FK506 in a calcineurin-dependent manner, is a cAMP response element (CRE)/activating transcription factor (ATF)-like element located 84 base pairs (bp) proximal to the transcriptional initiation site. Although transcription of the SNK gene was also regulated by tunicamycin, etoposide, or staurosporine, FK506 did not show any effects on these regulations. We recently reported that FK506 did not protect against apoptosis induced by these agents. These results indicate that the induction of SNK mRNA by thapsigargin in SH-SY5Y cells is regulated by FK506 via an inhibition of calcineurin at the transcriptional stage, and the transcriptional regulation of the SNK gene by FK506 was well correlated with the protective effect of the compound against apoptosis. Thus, transcriptional regulation of the SNK gene may be a biological marker for analysis of apoptosis of SH-SY5Y cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Proteínas Quinasas/genética , Tacrolimus/farmacología , Línea Celular Tumoral , Humanos , Proteínas Serina-Treonina Quinasas , Tapsigargina/farmacología , Activación Transcripcional/efectos de los fármacos
13.
Comp Med ; 52(5): 445-51, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12405638

RESUMEN

We have established safe and efficient methods for autologous hematopoietic stem cell (HSC) transplantation in cynomolgus monkeys (Macaca fascicularis) that include regimens of supportive care to ensure survival during hematopoietic reconstitution following otherwise lethal total body irradiation. Eleven young adult cynomolgus monkeys were studied. Bone marrow was aspirated from the ilium and/or tuber ischiae after administration of recombinant human stem cell factor (SCF) and granulocyte colony-stimulating factor (G-CSF). Using the immunomagnetic selection method, CD34+ cells were then isolated (90 to 95% pure) as a fraction containing HSCs. Just prior to transplantation, the animals received myeloablative total body irradiation-500 to 550 cGy daily for two days. The monkeys re-infused with CD34+ cells developed moderate to severe myelosuppression, with some animals requiring intravenous hyperalimentation, antibiotic administration, and blood transfusion. Hematopoiesis was restored in all animals after transplantation. It took 12 days, on average, until the peripheral white blood cell count reached more than 1,000 cells/microl. Up to two years after transplantation, signs of radiation-induced pneumonitis or other radiation-related disorders were not evident at the aforementioned dose of irradiation. This transplantation model will be useful for testing new approaches using HSCs for therapy of many diseases and will offer unique insights into the biology of these cells.


Asunto(s)
Investigación Biomédica , Trasplante de Células Madre Hematopoyéticas/veterinaria , Macaca fascicularis , Cuidados Posoperatorios/veterinaria , Trasplante Autólogo/métodos , Animales , Recuento de Células Sanguíneas/veterinaria , Médula Ósea , Femenino , Trasplante de Células Madre Hematopoyéticas/métodos , Masculino , Complicaciones Posoperatorias/veterinaria , Irradiación Corporal Total/veterinaria
14.
Eur J Pharmacol ; 645(1-3): 185-91, 2010 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-20655898

RESUMEN

Recent evidence suggests that the forkhead transcription factor Foxo1 plays an important role in the regulation of glucose and triglyceride metabolism at the gene transcription level for glucose-6 phosphatase (G6Pase), phosphoenolpyruvate carboxykinase (PEPCK), and apolipoprotein C-III (apoC-III). Here, we report on the pharmacological effects of the novel Foxo1 inhibitor AS1708727, which we identified by compound screening. Chronic treatment of diabetic db/db mice with AS1708727 for four days significantly reduced blood glucose and triglyceride levels with decrease of gene expression levels of hepatic G6Pase, PEPCK, and apoC-III. No reports have yet examined the influence of Foxo1 inhibitors on these pharmacological effects. In this study, we newly identified a Foxo1 inhibitor compound capable of exerting both an anti-hypertriglyceridemic and anti-hyperglycemic effect. These effects were dependent on maintaining a stable blood concentration of AS1708727 and achieving a high rate of compound transition to the liver. We also investigated the action mechanism of AS1708727 on gluconeogenesis in vitro and in vivo. The compound inhibited gene expression of key gluconeogenic molecules and suppressed gluconeogenesis in Fao hepatocyte cells in vitro. Further, in the pyruvate challenge study using db/db mice in vivo, AS1708727 suppressed increases in blood glucose level by inhibiting gluconeogenic gene expression. These results indicate that the novel Foxo1 inhibitor AS1708727 may exert anti-diabetic and anti-hypertriglyceridemic effects by improving blood glucose and triglyceride metabolism at the gene expression level, and may represent a new class of drugs useful for treating type 2 diabetes mellitus and hypertriglyceridemia.


Asunto(s)
Acetanilidas/farmacocinética , Glucemia/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Factores de Transcripción Forkhead/antagonistas & inhibidores , Hipoglucemiantes/farmacología , Hipolipemiantes/farmacología , Isoquinolinas/farmacocinética , Triglicéridos/sangre , Animales , Apolipoproteína C-III/metabolismo , Células Cultivadas , Proteína Forkhead Box O1 , Gluconeogénesis , Glucosa-6-Fosfatasa/metabolismo , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Hipertrigliceridemia/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Fosfoenolpiruvato Carboxiquinasa (GTP)/metabolismo
15.
J Control Release ; 126(1): 59-66, 2008 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-18055057

RESUMEN

RNA interference (RNAi) is a valuable tool for the validation of gene identification and functional genomics. Previously, it was reported that 6th generation dendritic poly(L-lysine) (KG6) transfected DNA into several cultivated cell lines with high efficiency and without any cytotoxic effects. In this study, the potential of KG6 to be an efficient siRNA carrier is investigated. KG6 showed effective knockdown of GAPDH with low cytotoxicity in combination with the weak-base amphiphilic peptide, Endo-Porter. In addition, the knockdown of PEPCK, which is the rate-limiting enzyme for gluconeogenesis, led to a reduction in glucose production in rat hepatoma H4IIEC3 cells. Knockdown of organic cation transporter 1 (OCT1), which is thought to be the gene that influences metformin action, was shown to successfully diminish the ability of metformin to inhibit gluconeogenesis in H4IIEC3 cells. In conclusion, using a combination of KG6 and Endo-Porter, a model system in which genes that influence metformin action can be identified was successfully constructed.


Asunto(s)
Dendrímeros/química , Portadores de Fármacos/química , Polilisina/química , Interferencia de ARN , ARN Interferente Pequeño/administración & dosificación , Animales , Proteínas de Transporte de Catecolaminas en la Membrana Plasmática/genética , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Gluconeogénesis/efectos de los fármacos , Gliceraldehído 3-Fosfato Deshidrogenasa (NADP+)/genética , Metformina/farmacología , Proteínas Serina-Treonina Quinasas/genética , ARN Interferente Pequeño/genética , Ratas , Transfección
16.
Exp Anim ; 54(5): 421-8, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16365519

RESUMEN

Hematopoietic stem cells in bone marrow can be mobilized into peripheral blood by cytokine administration. Cytokine-mobilized peripheral blood stem cells are of great use in clinical applications. We previously established a modified procedure for the collection of cytokine-mobilized peripheral blood cells from rhesus monkeys (Macaca mulata) using a commercially available apparatus originally developed for human subjects. In this study, we examined the efficacy and safety of this method with even smaller macaques, cynomolgus monkeys (Macaca fascicularis), which are equivalent to human newborns in body weight (mean = 3.3 kg). Using the manufacturer's unmodified protocol (n=6), one monkey died of cardiac failure and three developed severe anemia. In contrast, using our modified procedure (n=6), no such complication was observed in any animal. In addition, the harvested nuclear cell, mononuclear cell and CD34(+) cell counts were significantly higher with the modified method. The modified method should allow safe and efficient collection of cytokine-mobilized peripheral blood cells from non-human primates as small as human newborns in a non-invasive manner.


Asunto(s)
Células de la Médula Ósea/efectos de los fármacos , Factor Estimulante de Colonias de Granulocitos/farmacología , Movilización de Célula Madre Hematopoyética/métodos , Leucaféresis/métodos , Macaca fascicularis/fisiología , Factor de Células Madre/farmacología , Anemia/inducido químicamente , Animales , Recuento de Células Sanguíneas , Femenino , Células Madre Hematopoyéticas , Leucaféresis/instrumentación , Masculino
17.
J Neurochem ; 94(5): 1264-76, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15992370

RESUMEN

Nerve growth factor (NGF) plays a key role in the differentiation of neurons. In this study, we established three NGF-induced neurite-positive (NIN+) subclones that showed high responsiveness to NGF-induced neurite outgrowth and three NGF-induced neurite-negative (NIN-) subclones that abolished NGF-induced neurite outgrowth from parental SH-SY5Y cells, and analyzed differences in the NGF signaling cascade. The NIN+ subclones showed enhanced responsiveness to FK506-mediated neurite outgrowth as well. To clarify the mechanism behind the high frequency of NGF-induced neurite outgrowth, we investigated differences in NGF signaling cascade among subclones. Expression levels of the NGF receptor TrkA, and NGF-induced increases in mRNAs for the immediate-early genes (IEGs) c-fos and NGF inducible (NGFI) genes NGFI-A, NGFI-B and NGFI-C, were identical among subclones. Microarray analysis revealed that the NIN+ cell line showed a very different gene expression profile to the NIN- cell line, particularly in terms of axonal vesicle-related genes and growth cone guidance-related genes. Thus, the difference in NGF signaling cascade between the NIN+ and NIN- cell lines was demonstrated by the difference in gene expression profile. These differentially expressed genes might play a key role in neurite outgrowth of SH-SY5Y cells in a region downstream from the site of induction of IEGs, or in a novel NGF signaling cascade.


Asunto(s)
Perfilación de la Expresión Génica , Factor de Crecimiento Nervioso/farmacología , Neuritas/efectos de los fármacos , Neuroblastoma/fisiopatología , Línea Celular Tumoral , Células Clonales/metabolismo , Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Genes Inmediatos-Precoces , Humanos , Neuroblastoma/genética , Neuroblastoma/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Receptor trkA/metabolismo
18.
Bioorg Med Chem Lett ; 14(4): 847-50, 2004 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-15012979

RESUMEN

Monitoring of intracellular protein kinase activity is very important for fields involving diagnosis and drug screening. However, current methods, such as radiometry using (32)P, or ELISA, are laborious and time-consuming. We have developed high-throughput assay system of protein kinase activity using mass-tagged substrate peptide probes and mass spectrometry. This assay system can easily evaluate target kinase activity and will potentially be able to simultaneously profile many protein kinase activities.


Asunto(s)
Péptidos/química , Péptidos/metabolismo , Proteínas Quinasas/análisis , Proteínas Quinasas/metabolismo , Espectrometría de Masas/métodos , Proteínas Quinasas/química , Especificidad por Sustrato
19.
Biochem Biophys Res Commun ; 303(1): 170-6, 2003 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-12646182

RESUMEN

We previously developed "selective amplifier genes (SAGs)" which confer a growth advantage to transduced cells. The SAG is a chimeric gene encoding the G-CSF receptor (GCR) and the estrogen or tamoxifen (Tm) receptor and is able to expand transduced hematopoietic cells by treatment with estrogen or Tm. In the current study, we examined the in vitro efficacy of modified SAGs containing the thrombopoietin (TPO) receptor (c-Mpl) gene instead of GCR as a more potent signal generator. In addition, we constructed various mutant Mpl-type SAGs to abolish the responsiveness to endogenous TPO while retaining Tm-dependency. When Ba/F3 cells were retrovirally transduced with the Mpl-type SAGs, the cells showed Tm- and TPO-dependent growth even without IL-3. The Mpl-type SAGs induced more potent proliferation of Ba/F3 and cynomolgus CD34(+) cells than the GCR-type SAG. One mutant Mpl-type SAG (Delta GCRMplTmR) successfully lost the responsiveness to TPO without affecting the Tm-dependence.


Asunto(s)
Técnicas Genéticas , Células Madre Hematopoyéticas/citología , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Receptores de Citocinas/genética , Receptores de Citocinas/metabolismo , Animales , Antígenos CD34/biosíntesis , Sitios de Unión , Western Blotting , Células de la Médula Ósea/metabolismo , División Celular , Línea Celular , Dimerización , Relación Dosis-Respuesta a Droga , Citometría de Flujo , Eliminación de Gen , Terapia Genética , Humanos , Interleucina-3/metabolismo , Ratones , Mutación , Plásmidos/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Receptores de Trombopoyetina , Retroviridae/genética
20.
J Gene Med ; 6(1): 22-31, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14716674

RESUMEN

BACKGROUND: In vivo expansion of gene-modified cells would be a promising approach in the field of hematopoietic stem cell gene therapy. To this end, we previously developed a selective amplifier gene (SAG), a chimeric gene encoding the granulocyte colony-stimulating factor (G-CSF) receptor (GCR), as a growth-signal generator and the hormone-binding domain of the steroid receptor as a molecular switch. We have already reported that hematopoietic cells retrovirally transduced with the SAG can be expanded in a steroid-dependent manner in vitro and in vivo in mice and nonhuman primates. In this study, we have developed a new-generation SAG, in which the erythropoietin (EPO) receptor (EPOR) is utilized instead of the steroid receptor as a molecular switch. METHODS: Two EPO-driven SAGs were constructed, EPORGCR and EPORMpl, containing the GCR and c-Mpl as a signal generator, respectively. First, to compare the steroid-driven and EPO-driven SAGs, Ba/F3 cells were transduced with these SAGs. Next, to examine whether GCR or c-Mpl is the more suitable signal generator of the EPO-driven SAG, human cord blood CD34(+) cells were transduced with the two EPO-driven SAGs (EPORMpl and EPORGCR). Finally, we examined the in vivo efficacy of EPORMpl in mice. Irradiated mice were transplanted with EPORMpl-transduced bone marrow cells followed by administration of EPO. RESULTS: The EPO-driven SAGs were shown to induce more rapid and potent proliferation of Ba/F3 cells than the steroid-driven SAGs. The EPORMpl induced more efficient EPO-dependent proliferation of the human cord blood CD34(+) cells than the EPORGCR in terms of total CD34(+) cell, c-Kit(+) cell, and clonogenic progenitor cell (CFU-C) numbers. In the transplanted mice the transduced peripheral blood cells significantly increased in response to EPO. CONCLUSIONS: The new-generation SAGs, especially EPORMpl, are able to efficiently confer an EPO-dependent growth advantage on transduced hematopoietic cells in vitro and in vivo in mice.


Asunto(s)
Amplificación de Genes , Terapia Genética/métodos , Trasplante de Células Madre Hematopoyéticas , Receptores de Eritropoyetina/genética , Animales , Antígenos CD34/análisis , Antígenos CD34/inmunología , División Celular , Línea Celular , Vectores Genéticos , Ratones , Retroviridae/genética , Transducción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA