Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Transp Res Part A Policy Pract ; 161: 186-199, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35645469

RESUMEN

Coronavirus 2019 (COVID-19) and its variants are still spreading rapidly with deadly consequences and profound impacts on the global health and world economy. Without a suitable vaccine, mobility restriction has been the most effective method so far to prevent its spreading and avoid overwhelming the heath system of the affected country. The compartmental model SIR (or Susceptible, Infected, and Recovered) is the most popular mathematical model used to predict the course of the COVID-19 pandemic in order to plan the control actions and mobility restrictions against its spreading. A major limitation of this model in relation to modeling the spreading of COVID-19, and the mobility limitation strategy, is that the SIR model does not include mobility or take into account changes in mobility within its structure. This paper develops and tests a new hybrid SIR model; SIR-M which is integrated with an urban activity travel model to explore how it might improve the prediction of pandemic course and the testing of mobility limitation strategies in managing virus spread. The paper describes the enhanced methodology and tests a range of mobility limitation strategies on virus spread outcomes. Implications for policy and research futures are suggested.

3.
Mol Cancer Ther ; 13(3): 662-74, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24362465

RESUMEN

Evidence implicating dysregulation of the IRE1/XBP-1s arm of the unfolded protein response (UPR) in cancer pathogenesis (e.g., multiple myeloma) has prompted the development of IRE1 RNase inhibitors. Here, effects of cyclin-dependent kinase (CDK) inhibitor SCH727965 (dinaciclib) on the IRE1 arm of the UPR were examined in human leukemia and myeloma cells. Exposure of cells to extremely low (e.g., nmol/L) concentrations of SCH727965, a potent inhibitor of CDKs 1/2/5/9, diminished XBP-1s and Grp78 induction by the endoplasmic reticulum (ER) stress-inducers thapsigargin and tunicamycin, while sharply inducing cell death. SCH727965, in contrast to IRE1 RNase inhibitors, inhibited the UPR in association with attenuation of XBP-1s nuclear localization and accumulation rather than transcription, translation, or XBP-1 splicing. Notably, in human leukemia cells, CDK1 and 5 short hairpin RNA (shRNA) knockdown diminished Grp78 and XBP-1s upregulation while increasing thapsigargin lethality, arguing for a functional role for CDK1/5 in activation of the cytoprotective IRE1/XBP-1s arm of the UPR. In contrast, CDK9 or 2 inhibitors or shRNA knockdown failed to downregulate XBP-1s or Grp78. Furthermore, IRE1, XBP-1, or Grp78 knockdown significantly increased thapsigargin lethality, as observed with CDK1/5 inhibition/knockdown. Finally, SCH727965 diminished myeloma cell growth in vivo in association with XBP-1s downregulation. Together, these findings demonstrate that SCH727965 acts at extremely low concentrations to attenuate XBP-1s nuclear accumulation and Grp78 upregulation in response to ER stress inducers. They also highlight a link between specific components of the cell-cycle regulatory apparatus (e.g., CDK1/5) and the cytoprotective IRE1/XBP-1s/Grp78 arm of the UPR that may be exploited therapeutically in UPR-driven malignancies.


Asunto(s)
Compuestos Bicíclicos Heterocíclicos con Puentes/administración & dosificación , Proteína Quinasa CDC2/metabolismo , Quinasa 5 Dependiente de la Ciclina/metabolismo , Compuestos de Piridinio/administración & dosificación , Respuesta de Proteína Desplegada/efectos de los fármacos , Animales , Proteína Quinasa CDC2/genética , Línea Celular Tumoral , Óxidos N-Cíclicos , Quinasa 5 Dependiente de la Ciclina/genética , Proteínas de Unión al ADN/metabolismo , Chaperón BiP del Retículo Endoplásmico , Estrés del Retículo Endoplásmico/efectos de los fármacos , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Proteínas de Choque Térmico/metabolismo , Humanos , Indolizinas , Leucemia/tratamiento farmacológico , Leucemia/genética , Leucemia/patología , Empalme del ARN/genética , Factores de Transcripción del Factor Regulador X , Transducción de Señal/efectos de los fármacos , Tapsigargina/toxicidad , Factores de Transcripción/metabolismo , Proteína 1 de Unión a la X-Box
4.
Leuk Res ; 34(3): 379-86, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20117835

RESUMEN

Interactions between the multi-kinase inhibitor sorafenib and MEK1/2 inhibitors were investigated in DLBCL cells. Sorafenib (3-10 microM) triggered apoptosis in multiple GC and ABC lymphoma cells. Unexpectedly, sorafenib did not cause sustained ERK1/2 inactivation, and in SUDHL-6 and -16 cells, triggered ERK1/2 activation. Marginally toxic MEK1/2 inhibitor concentrations (5 microM PD184352) abrogated ERK1/2 activation in sorafenib-treated cells and synergistically potentiated apoptosis. MEK1 shRNA transfection also significantly increased sorafenib-mediated lethality. Sorafenib/PD184352 co-administration accelerated Mcl-1 down-regulation without up-regulating Bim(EL). Finally, ectopic Mcl-1 expression attenuated sorafenib/PD184352-mediated apoptosis. Together, these findings provide a theoretical basis for potentiating sorafenib anti-lymphoma activity by MEK1/2 inhibitors.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Linfoma de Células B Grandes Difuso/metabolismo , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Benzamidas/administración & dosificación , Bencenosulfonatos/administración & dosificación , Western Blotting , Línea Celular Tumoral , Inhibidores Enzimáticos/administración & dosificación , Quinasas MAP Reguladas por Señal Extracelular/efectos de los fármacos , Humanos , Quinasas Quinasa Quinasa PAM/efectos de los fármacos , Niacinamida/análogos & derivados , Compuestos de Fenilurea , Piridinas/administración & dosificación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sorafenib , Transfección
5.
Blood ; 112(3): 793-804, 2008 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-18505786

RESUMEN

Interactions between the dual Bcr/Abl and aurora kinase inhibitor MK-0457 and the histone deacetylase inhibitor vorinostat were examined in Bcr/Abl(+) leukemia cells, including those resistant to imatinib mesylate (IM), particularly those with the T315I mutation. Coadministration of vorinostat dramatically increased MK-0457 lethality in K562 and LAMA84 cells. Notably, the MK-0457/vorinostat regimen was highly active against primary CD34(+) chronic myelogenous leukemia (CML) cells and Ba/F3 cells bearing various Bcr/Abl mutations (ie, T315I, E255K, and M351T), as well as IM-resistant K562 cells exhibiting Bcr/Abl-independent, Lyn-dependent resistance. These events were associated with inactivation and down-regulation of wild-type (wt) and mutated Bcr/Abl (particularly T315I). Moreover, treatment with MK-0457 resulted in accumulation of cells with 4N or more DNA content, whereas coadministration of vorinostat markedly enhanced aurora kinase inhibition by MK-0457, and preferentially killed polyploid cells. Furthermore, vorinostat also interacted with a selective inhibitor of aurora kinase A and B to potentiate apoptosis without modifying Bcr/Abl activity. Finally, vorinostat markedly induced Bim expression, while blockade of Bim induction by siRNA dramatically diminished the capacity of this agent to potentiate MK-0457 lethality. Together, these findings indicate that vorinostat strikingly increases MK-0457 activity against IM-sensitive and -resistant CML cells through inactivation of Bcr/Abl and aurora kinases, as well as by induction of Bim.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Ácidos Hidroxámicos/farmacología , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Piperazinas/farmacología , Animales , Proteínas Reguladoras de la Apoptosis/antagonistas & inhibidores , Aurora Quinasa A , Aurora Quinasas , Proteína 11 Similar a Bcl2 , Benzamidas , Línea Celular Tumoral , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Proteínas de Fusión bcr-abl/antagonistas & inhibidores , Humanos , Mesilato de Imatinib , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Pirimidinas , Vorinostat
6.
Mol Pharmacol ; 72(3): 788-95, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17595328

RESUMEN

The effects of the multikinase inhibitor sorafenib (BAY 43-9006), an agent shown previously to induce apoptosis in human leukemia cells through inhibition of myeloid cell leukemia-1 (Mcl-1) translation, have been examined in Bcr/Abl(+) leukemia cells resistant to imatinib mesylate (IM). When administered at pharmacologically relevant concentrations (10-15 microM), sorafenib potently induced apoptosis in imatinib mesylate-resistant cells expressing high levels of Bcr/Abl, cells exhibiting a Bcr/Abl-independent, Lyn-dependent form of resistance, and CD34(+) cells obtained from imatinib-resistant patients. In addition, Ba/F3 cells expressing mutations rendering them resistant to IM (e.g., E255K, M351T) or to IM, dasatinib, and nilotinib (T315I) remained fully sensitive to sorafenib. Induction of apoptosis by sorafenib was associated with rapid and pronounced down-regulation of Mcl-1 and diminished signal transducer and activator of transcription (STAT) 5 phosphorylation and reporter activity but only very modest and delayed inactivation of the Bcr/Abl downstream target Crkl. Moreover, transfection with a constitutively active STAT5 construct partially but significantly protected cells from sorafenib lethality. Ba/F3 cells expressing Bcr/Abl mutations were as sensitive to sorafenib-induced Mcl-1 down-regulation and dephosphorylation of STAT5 and eukaryotic initiation factor 4E as wild-type cells. Finally, stable knockdown of Bcl-2-interacting mediator of cell death (Bim) with short hairpin RNA in K562 cells significantly diminished sorafenib lethality, arguing strongly for a functional role of this proapoptotic Bcl-2 family member in the lethality of this agent. Together, these findings suggest that sorafenib effectively induces apoptosis in highly imatinib-resistant chronic myelogenous leukemia cells, most likely by inhibiting or down-regulating targets (i.e., STAT5 and Mcl-1) downstream or independent of Bcr/Abl.


Asunto(s)
Apoptosis/efectos de los fármacos , Bencenosulfonatos/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , Antineoplásicos/farmacología , Benzamidas , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Resistencia a Antineoplásicos , Proteínas de Fusión bcr-abl/metabolismo , Humanos , Mesilato de Imatinib , Células K562 , Niacinamida/análogos & derivados , Compuestos de Fenilurea , Piperazinas/farmacología , Pirimidinas/farmacología , Factor de Transcripción STAT5/efectos de los fármacos , Sorafenib
7.
Mol Cell Biol ; 27(15): 5499-513, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17548474

RESUMEN

Sorafenib is a multikinase inhibitor that induces apoptosis in human leukemia and other malignant cells. Recently, we demonstrated that sorafenib diminishes Mcl-1 protein expression by inhibiting translation through a MEK1/2-ERK1/2 signaling-independent mechanism and that this phenomenon plays a key functional role in sorafenib-mediated lethality. Here, we report that inducible expression of constitutively active MEK1 fails to protect cells from sorafenib-mediated lethality, indicating that sorafenib-induced cell death is unrelated to MEK1/2-ERK1/2 pathway inactivation. Notably, treatment with sorafenib induced endoplasmic reticulum (ER) stress in human leukemia cells (U937) manifested by immediate cytosolic-calcium mobilization, GADD153 and GADD34 protein induction, PKR-like ER kinase (PERK) and eukaryotic initiation factor 2alpha (eIF2alpha) phosphorylation, XBP1 splicing, and a general reduction in protein synthesis as assessed by [35S]methionine incorporation. These events were accompanied by pronounced generation of reactive oxygen species through a mechanism dependent upon cytosolic-calcium mobilization and a significant decline in GRP78/Bip protein levels. Interestingly, enforced expression of IRE1alpha markedly reduced sorafenib-mediated apoptosis, whereas knockdown of IRE1alpha or XBP1, disruption of PERK activity, or inhibition of eIF2alpha phosphorylation enhanced sorafenib-mediated lethality. Finally, downregulation of caspase-2 or caspase-4 by small interfering RNA significantly diminished apoptosis induced by sorafenib. Together, these findings demonstrate that ER stress represents a central component of a MEK1/2-ERK1/2-independent cell death program triggered by sorafenib.


Asunto(s)
Bencenosulfonatos/farmacología , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/patología , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , Animales , Señalización del Calcio/efectos de los fármacos , Caspasa 2/metabolismo , Caspasas Iniciadoras/metabolismo , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Proteínas de Unión al ADN/metabolismo , Relación Dosis-Respuesta a Droga , Chaperón BiP del Retículo Endoplásmico , Activación Enzimática/efectos de los fármacos , Inducción Enzimática/efectos de los fármacos , Factor 2 Eucariótico de Iniciación/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Niacinamida/análogos & derivados , Proteínas Nucleares/metabolismo , Compuestos de Fenilurea , Fosforilación/efectos de los fármacos , Biosíntesis de Proteínas/efectos de los fármacos , Pliegue de Proteína , Empalme del ARN/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Factores de Transcripción del Factor Regulador X , Sorafenib , Factores de Transcripción , Proteína 1 de Unión a la X-Box , eIF-2 Quinasa/antagonistas & inhibidores , eIF-2 Quinasa/metabolismo
8.
Blood ; 109(9): 4006-15, 2007 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-17218385

RESUMEN

Interactions between MEK1/2 inhibitors and the dual Abl/Src kinase inhibitor dasatinib (BMS-354825) were examined in chronic myeloid leukemia (CML) cell lines and primary specimens. Cotreatment of K562 or LAMA cells with subtoxic or marginally toxic concentrations of PD184352 (or U0126) and dasatinib synergistically potentiated mitochondrial damage, caspase activation, and apoptosis. Similar interactions were observed in CD34(+) cells from one CML patient-derived but not in a normal human CD34(+) bone marrow cell specimen. These interactions were associated with multiple perturbations in survival signaling pathways, including inactivation of Bcr/Abl, STAT5, and ERK1/2; down-regulation of Bcl-x(L) and Mcl-1; and dephosphorylation/activation of Bim. They were also associated with BAX/BAK conformational change, mitochondrial dysfunction, and caspase activation. Bim knockdown by shRNA suppressed BAX and BAK conformational change and protected cells from dasatinib/PD184352 lethality. Conversely, K562 cells ectopically expressing Mcl-1 or Bcl-x(L) were significantly less susceptible to dasatinib/PD184352 toxicity. Notably, the dasatinib/PD184352 regimen was active against leukemic cells exhibiting various forms of imatinib mesylate resistance, including Bcr/Abl overexpression, Lyn activation, and several Bcr/Abl kinase domain mutations (eg, E255K, M351T), but not T315I. Together, these findings suggest that strategies combining dasatanib with MEK1/2 inhibitors warrant further investigation in Bcr/Abl(+) malignancies, particularly in the setting of imatinib mesylate-resistant disease.


Asunto(s)
Proteínas de Fusión bcr-abl/antagonistas & inhibidores , Leucemia Mielógena Crónica BCR-ABL Positiva/enzimología , MAP Quinasa Quinasa 1/antagonistas & inhibidores , MAP Quinasa Quinasa 2/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Tiazoles/farmacología , Benzamidas/agonistas , Benzamidas/farmacología , Butadienos/agonistas , Butadienos/farmacología , Dasatinib , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Proteínas de Fusión bcr-abl/metabolismo , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Regulación Leucémica de la Expresión Génica/genética , Humanos , Mesilato de Imatinib , Células K562 , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , MAP Quinasa Quinasa 1/metabolismo , MAP Quinasa Quinasa 2/metabolismo , MAP Quinasa Quinasa 2/farmacología , Nitrilos/agonistas , Nitrilos/farmacología , Piperazinas/farmacología , Piperazinas/uso terapéutico , Inhibidores de Proteínas Quinasas/agonistas , Pirimidinas/agonistas , Pirimidinas/uso terapéutico , Tiazoles/agonistas , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/metabolismo
9.
Clin Cancer Res ; 12(7 Pt 1): 2239-47, 2006 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-16609040

RESUMEN

PURPOSE: To characterize interactions between the heat shock protein 90 antagonist 17-dimethylaminoethylamino-17-demethoxygeldanamycin (DMAG) and the mitogen-activated protein kinase/extracellular signal-regulated kinase (ERK) kinase 1/2 inhibitor PD184352 in Bcr/abl(+) leukemia cells sensitive and resistant to imatinib mesylate. EXPERIMENTAL DESIGN: K562 and LAMA 84 cells were exposed to varying concentrations of DMAG and PD184352 for 48 hours; after which, mitochondrial integrity, caspase activation, and apoptosis were monitored. Parallel studies were done in imatinib mesylate-resistant cells, including BaF3 cells transfected with plasmids encoding clinically relevant Bcr/abl mutations conferring imatinib mesylate resistance (e.g., E255K, M351T, and T315I) and primary CD34(+) bone marrow cells from patients refractory to imatinib mesylate. RESULTS: Cotreatment of Bcr/abl(+) cells with minimally toxic concentrations of DMAG and PD184352 resulted in synergistic induction of mitochondrial injury (cytochrome c release and Bax conformational change), events associated with the pronounced and sustained inactivation of ERK1/2 accompanied by down-regulation of Bcl-x(L). Conversely, cells ectopically expressing Bcl-x(L) displayed significant protection against PD184352/DMAG-mediated lethality. This regimen effectively induced apoptosis in K562 cells overexpressing Bcr/abl, in BaF3 cells expressing various clinically relevant Bcr/abl mutations, and in primary CD34(+) cells from patients resistant to imatinib mesylate, but was relatively sparing of normal CD34(+) bone marrow cells. CONCLUSIONS: A regimen combining the heat shock protein 90 antagonist DMAG and the mitogen-activated protein kinase/ERK kinase 1/2 inhibitor potently induces apoptosis in Bcr/abl(+) cells, including those resistant to imatinib mesylate through various mechanisms including Bcr/abl kinase mutations, through a process that may involve sustained ERK1/2 inactivation and Bcl-x(L) down-regulation. This strategy warrants further attention in Bcr/abl(+) hematopoietic malignancies, particularly those resistant to Bcr/abl kinase inhibitors.


Asunto(s)
Antineoplásicos/farmacología , Benzamidas/farmacología , Proteínas de Fusión bcr-abl/metabolismo , Leucemia/tratamiento farmacológico , Piperazinas/farmacología , Pirimidinas/farmacología , Quinonas/farmacología , Rifabutina/análogos & derivados , Apoptosis/efectos de los fármacos , Benzoquinonas , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Resistencia a Antineoplásicos , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Inhibidores Enzimáticos/farmacología , Proteínas de Fusión bcr-abl/efectos de los fármacos , Humanos , Mesilato de Imatinib , Lactamas Macrocíclicas , Leucemia/metabolismo , Rifabutina/farmacología , Sensibilidad y Especificidad , Relación Estructura-Actividad , Factores de Tiempo , Células Tumorales Cultivadas
10.
Leuk Res ; 30(10): 1263-72, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16481037

RESUMEN

Effects of the tyrphostin adaphostin and bortezomib were examined in Bcr/Abl+ leukemia cell resistant to imatinib mesylate secondary to Bcr/Abl point mutations. Adaphostin was equally effective in inducing mitochondrial damage, caspase activation, JNK activation, and Raf-1, phospho-Stat3 and -Stat5 inactivation in mutant and wild-type cells, but differentially down-regulated phospho-Bcr/Abl. Adaphostin and bortezomib synergistically induced apoptosis in wild-type and mutant cells, including T315I mutants. Notably, adaphostin+/-bortezomib potently induced ROS and lethality in mutant cells, effects attenuated by the antioxidant NAC. These findings indicate that adaphostin+/-bortezomib circumvent imatinib resistance due to Bcr/Abl point mutations most likely through ROS generation.


Asunto(s)
Adamantano/análogos & derivados , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Ácidos Borónicos/farmacología , Proteínas de Fusión bcr-abl/genética , Células Madre Hematopoyéticas/efectos de los fármacos , Hidroquinonas/farmacología , Mutación , Estrés Oxidativo/efectos de los fármacos , Piperazinas/farmacología , Pirazinas/farmacología , Pirimidinas/farmacología , Adamantano/farmacología , Benzamidas , Bortezomib , Línea Celular Tumoral , Proteínas de Fusión bcr-abl/efectos de los fármacos , Células Madre Hematopoyéticas/patología , Células Madre Hematopoyéticas/fisiología , Humanos , Mesilato de Imatinib , Cinética , Mutación Puntual , Especies Reactivas de Oxígeno/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...