Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Alzheimers Dis ; 56(4): 1403-1417, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28157092

RESUMEN

The dyshomeostasis of transition metal ions, accumulation of amyloid-ß (Aß) senile plaques and neuroinflammatory response found in the brain of patients with Alzheimer's disease (AD) have been suggested to be involved in AD pathogenesis. Novel compounds capable of targeting metal-Aß species and neuroinflammation would be valuable. AD-35 is such a patented small-molecule compound derived from innovative modification of the chemical structure of donepezil. This compound could moderately inhibit acetylcholinesterase and metal-induced Aß aggregation in vitro and showed disassembly of Aß aggregates. The effects of AD-35 on cognitive impairments and neuroinflammatory changes caused by intracerebroventricular injection of Aß25-35 were studied in rats. Compared to sham group, Aß25-35 injection significantly led to learning and memory deficits, astrocyte activation, and pro-inflammatory cytokines releases (TNF-α and IL-1ß). Further studies indicated that the phosphorylation of extracellular signal-regulated kinase was involved in astrocyte activation and pro-inflammatory cytokines production. Oral administration of AD-35 could markedly attenuate Aß25-35 injection-induced astrocyte activation, pro-inflammatory cytokines TNF-α and IL-1ß release, and memory deficits. On the contrary, donepezil only showed inhibition of IL-1ß production, but failed to block astrocyte activation and TNF-α production. These results showed that AD-35 would be a novel multi-mechanism drug for the prevention and/or treatment of AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Encéfalo/efectos de los fármacos , Inhibidores de la Colinesterasa/farmacología , Disfunción Cognitiva/tratamiento farmacológico , Nootrópicos/farmacología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Astrocitos/patología , Encéfalo/metabolismo , Encéfalo/patología , Línea Celular Tumoral , Inhibidores de la Colinesterasa/química , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/patología , Modelos Animales de Enfermedad , Donepezilo , Humanos , Indanos/farmacología , Interleucina-1beta/metabolismo , Masculino , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/metabolismo , Trastornos de la Memoria/patología , Nootrópicos/química , Fragmentos de Péptidos , Piperidinas/farmacología , Agregación Patológica de Proteínas/tratamiento farmacológico , Agregación Patológica de Proteínas/metabolismo , Agregación Patológica de Proteínas/patología , Distribución Aleatoria , Ratas Sprague-Dawley , Factor de Necrosis Tumoral alfa/metabolismo
2.
J Clin Invest ; 126(5): 1759-72, 2016 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-27043283

RESUMEN

Multiple myeloma (MM) cells secrete osteoclastogenic factors that promote osteolytic lesions; however, the identity of these factors is largely unknown. Here, we performed a screen of human myeloma cells to identify pro-osteoclastogenic agents that could potentially serve as therapeutic targets for ameliorating MM-associated bone disease. We found that myeloma cells express high levels of the matrix metalloproteinase MMP-13 and determined that MMP-13 directly enhances osteoclast multinucleation and bone-resorptive activity by triggering upregulation of the cell fusogen DC-STAMP. Moreover, this effect was independent of the proteolytic activity of the enzyme. Further, in mouse xenograft models, silencing MMP-13 expression in myeloma cells inhibited the development of osteolytic lesions. In patient cohorts, MMP-13 expression was localized to BM-associated myeloma cells, while elevated MMP-13 serum levels were able to correctly predict the presence of active bone disease. Together, these data demonstrate that MMP-13 is critical for the development of osteolytic lesions in MM and that targeting the MMP-13 protein - rather than its catalytic activity - constitutes a potential approach to mitigating bone disease in affected patients.


Asunto(s)
Metaloproteinasa 13 de la Matriz/metabolismo , Mieloma Múltiple/enzimología , Proteínas de Neoplasias/metabolismo , Osteoclastos/enzimología , Osteólisis/enzimología , Animales , Fusión Celular , Femenino , Xenoinjertos , Humanos , Masculino , Metaloproteinasa 13 de la Matriz/genética , Ratones , Ratones Noqueados , Mieloma Múltiple/genética , Mieloma Múltiple/patología , Mieloma Múltiple/terapia , Proteínas de Neoplasias/genética , Trasplante de Neoplasias , Osteoclastos/patología , Osteólisis/genética , Osteólisis/patología , Osteólisis/terapia
3.
Mol Carcinog ; 54(12): 1796-806, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25640641

RESUMEN

Cannabinoid receptor-2 (CB2) is expressed dominantly in the immune system, especially on plasma cells. Cannabinergic ligands with CB2 selectivity emerge as a class of promising agents to treat CB2-expressing malignancies without psychotropic concerns. In this study, we found that CB2 but not CB1 was highly expressed in human multiple myeloma (MM) and primary CD138+ cells. A novel inverse agonist of CB2, phenylacetylamide but not CB1 inverse agonist SR141716, inhibited the proliferation of human MM cells (IC50 : 0.62 ∼ 2.5 µM) mediated by apoptosis induction, but exhibited minor cytotoxic effects on human normal mononuclear cells. CB2 gene silencing or pharmacological antagonism markedly attenuated phenylacetylamide's anti-MM effects. Phenylacetylamide triggered the expression of C/EBP homologous protein at the early treatment stage, followed by death receptor-5 upregulation, caspase activation, and ß-actin/tubulin degradation. Cell cycle related protein cdc25C and mitotic regulator Aurora A kinase were inactivated by phenylacetylamide treatment, leading to an increase in the ratio inactive/active cdc2 kinase. As a result, phosphorylation of CDK substrates was decreased, and the MM cell mitotic division was largely blocked by treatment. Importantly, phenylacetylamide could overcome the chemoresistance of MM cells against dexamethasone or melphalan. Thus, targeting CB2 may represent an attractive approach to treat cancers of immune origin.


Asunto(s)
Agonistas de Receptores de Cannabinoides/farmacología , Proliferación Celular/efectos de los fármacos , Citoesqueleto/efectos de los fármacos , Mitosis/efectos de los fármacos , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/genética , Receptor Cannabinoide CB2/genética , Actinas/genética , Apoptosis/efectos de los fármacos , Apoptosis/genética , Proteína Quinasa CDC2/genética , Caspasas/genética , Proteínas de Ciclo Celular/genética , Línea Celular Transformada , Proliferación Celular/genética , Citoesqueleto/genética , Silenciador del Gen/efectos de los fármacos , Humanos , Mitosis/genética , Receptor Cannabinoide CB2/agonistas , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética , Sindecano-1/genética , Tubulina (Proteína)/genética , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
4.
Biochem Biophys Res Commun ; 452(3): 334-9, 2014 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-25148941

RESUMEN

We performed molecular modeling and docking to predict a putative binding pocket and associated ligand-receptor interactions for human cannabinoid receptor 2 (CB2). Our data showed that two hydrophobic residues came in close contact with three structurally distinct CB2 ligands: CP-55,940, SR144528 and XIE95-26. Site-directed mutagenesis experiments and subsequent functional assays implicated the roles of Valine residue at position 3.32 (V113) and Leucine residue at position 5.41 (L192) in the ligand binding function and downstream signaling activities of the CB2 receptor. Four different point mutations were introduced to the wild type CB2 receptor: V113E, V113L, L192S and L192A. Our results showed that mutation of Val113 with a Glutamic acid and Leu192 with a Serine led to the complete loss of CB2 ligand binding as well as downstream signaling activities. Substitution of these residues with those that have similar hydrophobic side chains such as Leucine (V113L) and Alanine (L192A), however, allowed CB2 to retain both its ligand binding and signaling functions. Our modeling results validated by competition binding and site-directed mutagenesis experiments suggest that residues V113 and L192 play important roles in ligand binding and downstream signaling transduction of the CB2 receptor.


Asunto(s)
Canfanos/química , Agonistas de Receptores de Cannabinoides/química , Antagonistas de Receptores de Cannabinoides/química , Ciclohexanoles/química , Leucina/química , Pirazoles/química , Receptor Cannabinoide CB2/química , Valina/química , Animales , Sitios de Unión , Células CHO , Canfanos/metabolismo , Agonistas de Receptores de Cannabinoides/metabolismo , Antagonistas de Receptores de Cannabinoides/metabolismo , Cricetulus , AMP Cíclico/química , AMP Cíclico/metabolismo , Ciclohexanoles/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Leucina/genética , Ligandos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Mutagénesis Sitio-Dirigida , Unión Proteica , Pirazoles/metabolismo , Receptor Cannabinoide CB2/agonistas , Receptor Cannabinoide CB2/antagonistas & inhibidores , Receptor Cannabinoide CB2/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal , Valina/genética
5.
BMC Pharmacol Toxicol ; 15: 30, 2014 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-24913620

RESUMEN

BACKGROUND: Cannabinoid receptor 2 (CB2) is expressed predominantly in the immune system, particularly in plasma cells, raising the possibility that targeting the CB2 pathway could yield an immunomodulatory effect. Although the role of CB2 in mediating immunoglobulin class switching has been reported, the effects of targeting the CB2 pathway on immunoglobulin secretion per se remain unclear. METHODS: Human B cell line SKW 6.4, which is capable of differentiating into IgM-secreting cells once treated with human IL-6, was employed as the cell model. SKW 6.4 cells were incubated for 4 days with CB2 ligands plus IL-6 (100 U/ml). The amount of secreted IgM was determined by an ELISA. Cell proliferation was determined by the 3H-Thymidine incorporation assay. Signal molecules involved in the modulation of IgM secretion were examined by real-time RT-PCR and Western blot analyses or by using their specific inhibitors. RESULTS: We demonstrated that CB2 inverse agonists SR144528 and AM630, but not CB2 agonist HU308 or CB1 antagonist SR141716, effectively inhibited IL-6-induced secretion of soluble IgM without affecting cell proliferation as measured by thymidine uptake. SR144528 alone had no effects on the basal levels of IgM in the resting cells. These effects were receptor mediated, as pretreatment with CB2 agonist abrogated SR144528-mediated inhibition of IL-6 stimulated IgM secretion. Transcription factors relevant to B cell differentiation, Bcl-6 and PAX5, as well as the protein kinase STAT3 pathway were involved in the inhibition of IL-6-induced IgM by SR144528. CONCLUSIONS: These results uncover a novel function of CB2 antagonists and suggest that CB2 ligands may be potential modulators of immunoglobulin secretion.


Asunto(s)
Inmunoglobulina M/metabolismo , Interleucina-6/farmacología , Receptor Cannabinoide CB2/antagonistas & inhibidores , Canfanos/farmacología , Cannabinoides/farmacología , Línea Celular , Proliferación Celular/efectos de los fármacos , Proteínas de Unión al ADN/metabolismo , Humanos , Indoles/farmacología , Factor de Transcripción PAX5/metabolismo , Piperidinas/farmacología , Proteínas Proto-Oncogénicas c-bcl-6 , Pirazoles/farmacología , Receptor Cannabinoide CB1/antagonistas & inhibidores , Receptor Cannabinoide CB2/agonistas , Receptor Cannabinoide CB2/metabolismo , Rimonabant , Factor de Transcripción STAT3/metabolismo
6.
J Med Chem ; 56(5): 2045-58, 2013 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-23406429

RESUMEN

Cannabinoid receptors have gained increasing attention as drug targets for developing potential therapeutic ligands. Here, we report the discovery and optimization of triaryl sulfonamides as a novel series possessing significant CB2 receptor affinity and selectivity. Four sets of triaryl ligands were designed and synthesized for further structural modifications and led to the identification of eight compounds as potent and selective CB2 inverse agonists with high binding affinity (CB2K(i) < 10 nM). Especially, compound 57 exhibited the strongest binding affinity on the CB2 receptor (CB2K(i) of 0.5 nM) and the best selectivity over the CB1 receptor (selectivity index of 2594). Importantly, 57 also showed potent inhibitory activity on osteoclast formation, and it was confirmed by a cell viability assay that the inhibition effects were not derived from the cytotoxicity. Finally, 3D QSAR studies confirmed our SAR findings that three bulky groups play an important role for CB2 receptor binding affinity.


Asunto(s)
Agonistas de Receptores de Cannabinoides/síntesis química , Osteoclastos/efectos de los fármacos , Receptor Cannabinoide CB2/agonistas , Receptor Cannabinoide CB2/metabolismo , Sulfonamidas/farmacología , Animales , Bioensayo , Células CHO , Agonistas de Receptores de Cannabinoides/farmacología , Cricetinae , Ligandos , Macrófagos/efectos de los fármacos , Ratones , Relación Estructura-Actividad Cuantitativa
7.
ACS Med Chem Lett ; 4(4): 387-392, 2013 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-24729834

RESUMEN

An extensive exploration of the SAR of a trisubstituted sulfonamides series led to the identification of 39, which is a potent and selective CB2 receptor inverse agonist (CB2Ki = 5.4 nM, and CB1Ki = 500 nM). The functional properties measured by cAMP assays indicated that the selected compounds were CB2 inverse agonists with high potency values (34, EC50 = 8.2 nM, and 39, EC50 = 2.5 nM). Furthermore, an osteoclastogenesis bioassay indicated that trisubstituted sulfonamide compounds showed great inhibition of osteoclast formation.

8.
J Med Chem ; 55(22): 9973-87, 2012 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-23072339

RESUMEN

N,N'-((4-(Dimethylamino)phenyl)methylene)bis(2-phenylacetamide) was discovered by using 3D pharmacophore database searches and was biologically confirmed as a new class of CB(2) inverse agonists. Subsequently, 52 derivatives were designed and synthesized through lead chemistry optimization by modifying the rings A-C and the core structure in further SAR studies. Five compounds were developed and also confirmed as CB(2) inverse agonists with the highest CB(2) binding affinity (CB(2)K(i) of 22-85 nM, EC(50) of 4-28 nM) and best selectivity (CB(1)/CB(2) of 235- to 909-fold). Furthermore, osteoclastogenesis bioassay indicated that PAM compounds showed great inhibition of osteoclast formation. Especially, compound 26 showed 72% inhibition activity even at the low concentration of 0.1 µM. The cytotoxicity assay suggested that the inhibition of PAM compounds on osteoclastogenesis did not result from its cytotoxicity. Therefore, these PAM derivatives could be used as potential leads for the development of a new type of antiosteoporosis agent.


Asunto(s)
Bencenoacetamidas/farmacología , Médula Ósea/efectos de los fármacos , Osteoclastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Receptor Cannabinoide CB2/agonistas , Alquilación/efectos de los fármacos , Animales , Bencenoacetamidas/química , Unión Competitiva , Células CHO , Muerte Celular , Células Cultivadas , Cricetinae , AMP Cíclico/metabolismo , Ensayos Analíticos de Alto Rendimiento , Humanos , Modelos Moleculares , Estructura Molecular , Osteoclastos/citología , Receptor Cannabinoide CB2/metabolismo , Relación Estructura-Actividad
9.
Protein Expr Purif ; 83(2): 128-34, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22406258

RESUMEN

G protein coupled receptors (GPCRs) are key players in signal recognition and cellular communication making them important therapeutic targets. Large-scale production of these membrane proteins in their native form is crucial for understanding their mechanism of action and target-based drug design. Here we report the overexpression system for a GPCR, the cannabinoid receptor subtype 2 (CB2), in Escherichia coli C43(DE3) facilitated by two fusion partners: Mistic, an integral membrane protein expression enhancer at the N-terminal, and TarCF, a C-terminal fragment of the bacterial chemosensory transducer Tar at the C-terminal of the CB2 open reading frame region. Multiple histidine tags were added on both ends of the fusion protein to facilitate purification. Using individual and combined fusion partners, we found that CB2 fusion protein expression was maximized only when both partners were used. Variable growth and induction conditions were conducted to determine and optimize protein expression. More importantly, this fusion protein Mistic-CB2-TarCF can localize into the E. coli membrane and exhibit functional binding activities with known CB2 ligands including CP55,940, WIN55,212-2 and SR144,528. These results indicate that this novel expression and purification system provides us with a promising strategy for the preparation of biologically active GPCRs, as well as general application for the preparation of membrane-bound proteins using the two new fusion partners described.


Asunto(s)
Proteínas de Escherichia coli/metabolismo , Escherichia coli/genética , Receptor Cannabinoide CB2/metabolismo , Receptores de Superficie Celular/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Membrana Celular/metabolismo , Escherichia coli/metabolismo , Proteínas de Escherichia coli/biosíntesis , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Histidina/genética , Histidina/metabolismo , Humanos , Modelos Moleculares , Unión Proteica , Receptor Cannabinoide CB2/biosíntesis , Receptor Cannabinoide CB2/química , Receptor Cannabinoide CB2/genética , Receptores de Superficie Celular/biosíntesis , Receptores de Superficie Celular/química , Receptores de Superficie Celular/genética , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética
10.
Free Radic Biol Med ; 52(5): 889-97, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22226830

RESUMEN

Apoptosis requires tightly regulated cell death pathways. The signaling pathways that trigger a cell to undergo apoptosis after UV radiation are cell type specific and are currently being defined. Here, we have used pharmacological and genetic tools to demonstrate the decisive part of the mitochondrial pathway in UVC-induced apoptosis in mouse embryo fibroblasts (MEFs). UVC-induced apoptosis proceeded independent of the activation of death receptor components. In contrast, soon after UV radiation, MAPK activation and generation of reactive oxygen species (ROS) increased, followed by a decline in mitochondrial membrane potential (MMP) and cytochrome c release, as well as activation of caspase-9 and -3 and the upregulation of p47-phox. Deficiency of apaf-1, a critical member of the apoptosome, dramatically abolished all the UV-induced signal deterioration and cell death. In parallel, UVC-induced apoptosis was largely attenuated by either DN-caspase-9 or Bcl-X(L) overexpression. Pretreatment of cells with N-acetylcysteine or catalase but not Tempol decreased UVC-induced MAPK activation and apoptosis. Inhibition of JNK and caspase attenuated p47-phox upregulation. Altogether, we have for the first time demonstrated the critical role of Apaf-1 in the regulation of MAPK, ROS, and MMP in UVC-radiated MEFs and propose that the amplification feedback loop among mitochondrial signal molecules culminates in the demise of the cell.


Asunto(s)
Apoptosis/efectos de la radiación , Factor Apoptótico 1 Activador de Proteasas/deficiencia , Fibroblastos/fisiología , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Rayos Ultravioleta , Animales , Caspasa 8/metabolismo , Proteínas de Ciclo Celular/metabolismo , División Celular/efectos de la radiación , Forma del Núcleo Celular/efectos de la radiación , Células Cultivadas , Activación Enzimática , Proteína de Dominio de Muerte Asociada a Fas/genética , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Fibroblastos/efectos de la radiación , Sistema de Señalización de MAP Quinasas , Potencial de la Membrana Mitocondrial , Ratones
11.
Mol Cancer Ther ; 10(10): 1886-96, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21825007

RESUMEN

We identified nocodazole as a potent antimyeloma drug from a drug screening library provided by the Multiple Myeloma Research Foundation. Nocodazole is a benzimidazole that was originally categorized as a broad-spectrum anthelmintic drug with antineoplastic properties. We found that nocodazole inhibited growth and induced apoptosis of primary and multiresistant multiple myeloma cells cultured alone and in the presence of bone marrow stromal cells. Nocodazole caused cell-cycle prophase and prometaphase arrest accompanied by microtubular network disarray. Signaling studies indicated that increased expression of Bim protein and reduced X-linked inhibitor of apoptosis protein and Mcl-1(L) levels were involved in nocodazole-induced apoptosis. Further investigation showed Bcl-2 phosphorylation as a critical mediator of cell death, triggered by the activation of c-jun-NH(2) kinase (JNK) instead of p38 kinase or extracellular signal-regulated kinases. Treatment with JNK inhibitor decreased Bcl-2 phosphorylation and subsequently reduced nocodazole-induced cell death. Nocodazole combined with dexamethasone significantly inhibited myeloma tumor growth and prolonged survival in a human xenograft mouse model. Our studies show that nocodazole has potent antimyeloma activity and that targeting the microtubular network might be a promising new treatment approach for multiple myeloma.


Asunto(s)
Microtúbulos/efectos de los fármacos , Mieloma Múltiple/tratamiento farmacológico , Nocodazol/farmacología , Animales , Antineoplásicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Ciclo Celular/efectos de los fármacos , Procesos de Crecimiento Celular/efectos de los fármacos , Línea Celular Tumoral , Dexametasona/administración & dosificación , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Humanos , MAP Quinasa Quinasa 4/antagonistas & inhibidores , MAP Quinasa Quinasa 4/metabolismo , Ratones , Mieloma Múltiple/patología , Nocodazol/administración & dosificación , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Distribución Aleatoria , Moduladores de Tubulina/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Leuk Res ; 34(7): 917-24, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20116850

RESUMEN

We used a novel high-throughput drug screening assay, based on Luminex technology, to identify anti-myeloma agents capable of inhibiting cytokines and growth factors essential for multiple myeloma (MM) from a chemical library of 1120 compounds provided by MMRF. Tetracycline derivatives inhibited MM cell proliferation and osteoclast activating factors without obvious effect on cell viability. Steroid compounds specifically decreased angiogenesis-related factors, but stimulated osteoclast activating factors. Antihelmintic drugs potently inhibited cytokines and were cytotoxic. The screen identified potential candidates with potent anti-MM properties that need further investigation.


Asunto(s)
Antineoplásicos/farmacología , Citocinas/antagonistas & inhibidores , Ensayos de Selección de Medicamentos Antitumorales/métodos , Inhibidores de Crecimiento/farmacología , Ensayos Analíticos de Alto Rendimiento/métodos , Inmunoensayo/métodos , Mieloma Múltiple/tratamiento farmacológico , Corticoesteroides/farmacología , Antihelmínticos/farmacología , Antiinfecciosos/farmacología , Antineoplásicos/aislamiento & purificación , Apoptosis/efectos de los fármacos , Linfocitos B/efectos de los fármacos , Linfocitos B/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Glicósidos Cardíacos/farmacología , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral/efectos de los fármacos , Línea Celular Tumoral/metabolismo , Citocinas/metabolismo , Inhibidores de Crecimiento/aislamiento & purificación , Humanos , Microesferas , Osteoclastos/citología
13.
J Agric Food Chem ; 58(7): 3957-64, 2010 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-20025272

RESUMEN

The present study was performed to determine whether anthocyanins could trigger different modes of cell death in different cancers. It was found that whereas cyanidin-3-rutinoside and delphinidin could induce apoptosis in leukemia cells, they caused growth retardation in hepatocellular carcinoma cells (HCC), which was accompanied with a significant cellular vacuolization. The latter was likely caused by macroautophagy and was completely suppressed by 3-methyladenine, an inhibitor of class III phosphoinositide 3-kinase that is important for autophagy activation, and by bafilomycin A1, which blocks lysosomal degradation. Delphinidin induced significant lipidation of LC3, an indication of macroautophagy, which was also suppressed by 3-methyladenine. Macroautophagy was required for the survival of delphinidin-treated HCC cells as inhibition with 3-methyladenine led to massive necrosis without caspase activation. Thus, anthocyanins could induce different modes of cell death for different cancers. Furthermore, anthocyanins could be used in combination with a macroautophagy inhibitor for treating cancers such as HCC.


Asunto(s)
Antocianinas/farmacología , Autofagia/efectos de los fármacos , Carcinoma Hepatocelular/fisiopatología , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Regulación hacia Abajo , Humanos , Necrosis
14.
Mol Cancer Ther ; 7(6): 1494-505, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18566220

RESUMEN

Histone deacetylase inhibitors have emerged as promising anticancer drugs. Using an unbiased ultrahigh throughput screening system, a novel mercaptoketone-based histone deacetylase inhibitor series was identified that was optimized to the lead compound, KD5170. KD5170 inhibited the proliferation of myeloma cell lines and the viability of CD138(+) primary myeloma cells by induction of apoptosis, accompanied by an increase of acetylation of histones and activation of caspase-3, caspase-8, and caspase-9. Treatment with KD5170 caused a loss of mitochondrial membrane potential resulting in release of apoptogenic factors such as cytochrome c, Smac, and apoptosis-inducing factor. Furthermore, KD5170 induced oxidative stress and oxidative DNA damage in myeloma cells as evidenced by the up-regulation of heme oxygenase-1 and H2A.X phosphorylation. Combination of KD5170 with proteasome inhibitor bortezomib or tumor necrosis factor-related apoptosis-inducing ligand synergistically enhanced the antimyeloma activity. We further found that resistance of myeloma cells to KD5170 was associated with activation of the extracellular signal-regulated kinase/mitogen-activated protein kinase pathway under treatment with KD5170. Pretreatment with the mitogen-activated protein kinase inhibitor U0126 restored sensitivity to KD5170, suggesting that the combination of KD5170 with U0126 could overcome drug resistance. Growth of myeloma tumor xenografts in KD5170-treated nude mice was significantly inhibited and survival was prolonged. Histone acetylation was increased in spleen and tumor tissues of animals treated with KD5170. Our data indicate that KD5170 has potent antimyeloma activity in vitro and in vivo, which is mediated by DNA damage and mitochondrial signaling and subsequent induction of apoptosis.


Asunto(s)
Daño del ADN , Inhibidores Enzimáticos/farmacología , Inhibidores de Histona Desacetilasas , Cetonas/farmacología , Mitocondrias/metabolismo , Mieloma Múltiple/enzimología , Piridinas/farmacología , Transducción de Señal/efectos de los fármacos , Sulfonamidas/farmacología , Acetilación/efectos de los fármacos , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Proteínas Reguladoras de la Apoptosis/metabolismo , Ácidos Borónicos/farmacología , Bortezomib , Caspasas/metabolismo , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de los fármacos , Sinergismo Farmacológico , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/química , Histonas/metabolismo , Humanos , Cetonas/química , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Mitocondrias/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Pirazinas/farmacología , Piridinas/química , Sulfonamidas/química , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto , Proteína X Asociada a bcl-2/metabolismo
15.
Drug News Perspect ; 20(7): 431-5, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17992265

RESUMEN

Multiple myeloma (MM) is a hematologic malignancy characterized by dysregulated proliferation of plasma cells and increased osteoclast activity that results in bone destruction and lytic lesions. Therefore, novel treatment modalities targeting both myeloma cell and osteoclast function may dramatically improve MM patient outcome. As an R-enantiomer of the cyclooxygenase (COX) inhibitor etodolac, SDX-101 has shown favorable antimyeloma effects. More recently, another structural analogue of etodolac, SDX-308, has displayed in vitro cytotoxic activity on tumor lines and in vivo antitumor efficacy in mice. SDX-308 has high antimyeloma activity and shows synergism in combination with other drugs for the treatment of chronic lymphocytic leukemia (CLL). In addition SDX-308 inhibits osteoclast (OCL) formation resulting in complete abrogation of bone resorption. Therefore, SDX-308 might be an attractive drug for the treatment of diseases with increased OCL activity, such as osteolytic lesions in multiple myeloma and metastatic carcinomas as well as osteoporosis. In the present review, we discuss SDX-308 as a new therapeutic candidate for the treatment of MM and diseases with increased osteoclast activity.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Enfermedades Óseas/tratamiento farmacológico , Etodolaco/uso terapéutico , Compuestos Heterocíclicos con 3 Anillos/uso terapéutico , Mieloma Múltiple/tratamiento farmacológico , Osteoclastos/efectos de los fármacos , Antiinflamatorios no Esteroideos/farmacología , Antineoplásicos/uso terapéutico , Quimioterapia/tendencias , Compuestos Heterocíclicos con 3 Anillos/farmacología , Humanos , Mieloma Múltiple/fisiopatología , Factor de Necrosis Tumoral alfa/efectos de los fármacos
16.
Br J Haematol ; 139(3): 385-97, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17910628

RESUMEN

Clinical trials have shown the high anti-myeloma activity of the proteasome inhibitor bortezomib. The present study examined the activity of bortezomib combined with PXD101, a histone deacetylase inhibitor, against multiple myeloma (MM) and osteoclastogenesis. Treatment of myeloma cell lines with combinations of bortezomib and PXD101 led to synergistic inhibition of proliferation and induction of cell death. The combination significantly decreased the viability of primary human CD138(+) myeloma cells but not of bone marrow mononuclear cells. Further studies showed a dose-dependent activation of caspases-3, -8 and -9 and nuclear fragmentation in myeloma cells. Bortezomib/PXD101 treatment markedly triggered reactive oxygen species (ROS) generation that was accompanied by p53, H2A.X and p38-mitogen-activated protein kinase phosphorylation. ROS generation could be blocked by the free radical scavenger N-acetyl-L-cysteine. The combination of bortezomib and PXD101 also resulted in synergistic inhibition of osteoclast formation. In conclusion, bortezomib and PXD101 have different molecular targets. The combination induces cell death in myeloma cells via ROS-mediated DNA damage and also inhibits osteoclastogenesis. Therefore, this study provides the rationale for the clinical evaluation of bortezomib combined with PXD101 in patients with MM.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Daño del ADN , Mieloma Múltiple/patología , Estrés Oxidativo/efectos de los fármacos , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteína 11 Similar a Bcl2 , Células de la Médula Ósea/efectos de los fármacos , Ácidos Borónicos/administración & dosificación , Ácidos Borónicos/farmacología , Bortezomib , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , ADN de Neoplasias/genética , Evaluación Preclínica de Medicamentos , Sinergismo Farmacológico , Inhibidores Enzimáticos/farmacología , Inhibidores de Histona Desacetilasas , Humanos , Ácidos Hidroxámicos/administración & dosificación , Ácidos Hidroxámicos/farmacología , Proteínas de la Membrana/metabolismo , Mieloma Múltiple/genética , Mieloma Múltiple/metabolismo , Proteínas de Neoplasias/metabolismo , Osteogénesis/efectos de los fármacos , Proteínas Proto-Oncogénicas/metabolismo , Pirazinas/administración & dosificación , Pirazinas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Sulfonamidas
17.
Planta Med ; 73(5): 451-60, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17394101

RESUMEN

Fruit of deerberry [Vaccinium stamineum L.] were evaluated for their antioxidant capacity and anticancer properties in JB6 P (+) mouse epidermal cells, human lung and leukemia cells. Deerberries contain potent free radical scavenging activities. Pretreatment of JB6 P (+) mouse epidermal cells with deerberry fruit extracts produced an inhibition on the activation of activator protein-1 (AP-1) and nuclear factor-kappaB (NF-kappaB) induced by either 12- O-tetradecanoylphorbol 13-acetate (TPA) or ultraviolet-B (UVB). Deerberry fruit extracts also blocked TPA- or UVB-induced phosphorylation of ERKs and MEK 1/2, two upstream regulators of AP-1 and inhibited proliferation of human leukemia HL-60 cancer cells and human lung epithelial cancer A549 cells and induced apoptosis of HL-60 cells. These results suggest that the inhibition of TPA- or UVB-induced AP-1 and NF-kappaB activity, inhibition of HL-60 cells and cancer A549 cells proliferation and induction of apoptotic in human leukemia HL-60 cancer cells may be mediated through the ERKs and MEK 1/2 signal pathway.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Antioxidantes/farmacología , Extractos Vegetales/farmacología , Vaccinium/química , Animales , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/aislamiento & purificación , Antioxidantes/química , Antioxidantes/aislamiento & purificación , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cromatografía Líquida de Alta Presión , Activación Enzimática/efectos de los fármacos , Activación Enzimática/efectos de la radiación , Flavonoides/química , Flavonoides/aislamiento & purificación , Frutas/química , Humanos , Leucemia/enzimología , Leucemia/metabolismo , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/metabolismo , Ratones , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/antagonistas & inhibidores , Fosforilación/efectos de los fármacos , Fosforilación/efectos de la radiación , Extractos Vegetales/química , Extractos Vegetales/aislamiento & purificación , Especies Reactivas de Oxígeno/metabolismo , Acetato de Tetradecanoilforbol/farmacología , Factor de Transcripción AP-1/antagonistas & inhibidores , Rayos Ultravioleta
18.
J Biol Chem ; 282(18): 13468-76, 2007 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-17360708

RESUMEN

Anthocyanins are a group of naturally occurring phenolic compounds widely available in fruits and vegetables in human diets. They have broad biological activities including anti-mutagenesis and anticarcinogenesis, which are generally attributed to their antioxidant activities. We studied the effects and the mechanisms of the most common type of anthocyanins, cyanidin-3-rutinoside, in several leukemia and lymphoma cell lines. We found that cyanidin-3-rutinoside extracted and purified from the black raspberry cultivar Jewel induced apoptosis in HL-60 cells in a dose- and time-dependent manner. Paradoxically, this compound induced the accumulation of peroxides, which are involved in the induction of apoptosis in HL-60 cells. In addition, cyanidin-3-rutinoside treatment resulted in reactive oxygen species (ROS)-dependent activation of p38 MAPK and JNK, which contributed to cell death by activating the mitochondrial pathway mediated by Bim. Down-regulation of Bim or overexpression of Bcl-2 or Bcl-x(L) considerably blocked apoptosis. Notably, cyanidin-3-rutinoside treatment did not lead to increased ROS accumulation in normal human peripheral blood mononuclear cells and had no cytotoxic effects on these cells. These results indicate that cyanidin-3-rutinoside has the potential to be used in leukemia therapy with the advantages of being widely available and selective against tumors.


Asunto(s)
Antocianinas/farmacología , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Leucemia/metabolismo , Estrés Oxidativo/efectos de los fármacos , Extractos Vegetales/farmacología , Antocianinas/química , Antioxidantes/química , Proteínas Reguladoras de la Apoptosis/biosíntesis , Proteína 11 Similar a Bcl2 , Regulación hacia Abajo/efectos de los fármacos , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Células HL-60 , Humanos , Leucemia/tratamiento farmacológico , Proteínas de la Membrana/biosíntesis , Extractos Vegetales/química , Proteínas Proto-Oncogénicas/biosíntesis , Especies Reactivas de Oxígeno/metabolismo , Rosaceae/química , Proteína bcl-X/biosíntesis
19.
Blood ; 109(5): 2130-8, 2007 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-17095620

RESUMEN

Multiple myeloma is characterized by increased osteoclast activity that results in bone destruction and lytic lesions. With the prolonged overall patient survival achieved by new treatment modalities, additional drugs are required to inhibit bone destruction. We focused on a novel and more potent structural analog of the nonsteroidal anti-inflammatory drug etodolac, known as SDX-308, and its effects on osteoclastogenesis and multiple myeloma cells. SDX-101 is another structural analog of etodolac that is already used in clinical trials for the treatment of B-cell chronic lymphocytic leukemia (B-CLL). Compared with SDX-101, a 10-fold lower concentration of SDX-308 induced potent (60%-80%) inhibition of osteoclast formation, and a 10- to 100-fold lower concentration inhibited multiple myeloma cell proliferation. Bone resorption was completely inhibited by SDX-308, as determined in dentin-based bone resorption assays. SDX-308 decreased constitutive and RANKL-stimulated NF-kappaB activation and osteoclast formation in an osteoclast cellular model, RAW 264.7. SDX-308 effectively suppressed TNF-alpha-induced IKK-gamma and IkappaB-alpha phosphorylation and degradation and subsequent NF-kappaB activation in human multiple myeloma cells. These results indicate that SDX-308 effectively inhibits multiple myeloma cell proliferation and osteoclast activity, potentially by controlling NF-kappaB activation signaling. We propose that SDX-308 is a promising therapeutic candidate to inhibit multiple myeloma growth and osteoclast activity and that it should receive attention for further study.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Diferenciación Celular/efectos de los fármacos , Compuestos Heterocíclicos con 3 Anillos/farmacología , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , FN-kappa B/antagonistas & inhibidores , Osteoclastos/efectos de los fármacos , Animales , Antiinflamatorios no Esteroideos/química , Antiinflamatorios no Esteroideos/toxicidad , Resorción Ósea/metabolismo , Proliferación Celular/efectos de los fármacos , Compuestos Heterocíclicos con 3 Anillos/química , Compuestos Heterocíclicos con 3 Anillos/toxicidad , Humanos , Ratones , FN-kappa B/metabolismo , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Osteoclastos/citología , Osteoclastos/metabolismo , Ligando RANK/farmacología , Transducción de Señal , Células Tumorales Cultivadas
20.
J Toxicol Environ Health A ; 69(16): 1511-26, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16854782

RESUMEN

It is well known that ozone (O3), a potent reactive oxidant and air pollutant, induces respiratory inflammation and hyperresponsiveness upon inhalation. It was previously shown that O3 exposure (0.6 ppm, 10 h/day for 15 days) not only results in local bronchial inflammation, but also affects the nervous system and thymocyte proliferation, and places mice under oxidative stress. In the present study, data showed that O3 exposure could impair both the natural killer (NK) cell activity and the proliferation potential of spleen T cells to a specific antigen stimulus. Immunological function assays indicated that O3 exposure attenuated the proliferation of spleen mononuclear cells induced by concanavalin A and decreased CD4+ and CD28+ lymphocyte subsets. However, supplementation with natural antioxidants protected mice from O3-induced dysfunction of splenocyte proliferation. Meanwhile, O3 exposure resulted in a decline of mitogen-induced IL-2 production in splenocytes. It was also found that O3 exposure dramatically enhanced the proliferation of CD4-CD8- thymocytes stimulated by recombinant mouse interleukin-7 (rmIL-7), which is usually observed during the mammal aging process. Taken together, data conclude that short-term repetitive O3 exposure damages both innate and acquired immunity via altering the lymphocyte subset and cytokine profile, and via impact on thymocyte early development. O3-induced oxidative damage is one of the key factors leading to immune dysfunction in this mouse model.


Asunto(s)
Formación de Anticuerpos/efectos de los fármacos , Oxidantes Fotoquímicos/toxicidad , Ozono/toxicidad , Animales , Antígenos CD4 , Antígenos CD8 , Proliferación Celular , Células Asesinas Naturales , Ratones , Ratones Endogámicos BALB C , Estrés Oxidativo , Bazo/citología , Timo/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...