Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Cell Tissue Res ; 392(3): 733-743, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36988705

RESUMEN

The non-receptor tyrosine kinase Src plays a key role in cell division, migration, adhesion, and survival. Src is overactivated in several cancers, where it transmits signals that promote cell survival, mitosis, and other important cancer hallmarks. Src is therefore a promising target in cancer therapy, but the underlying mechanisms are still uncertain. Here we show that Src is highly conserved across different species. Src expression increases during mitosis and is localized to the chromosomal passenger complex. Knockdown or inhibition of Src induces multipolar spindle formation, resulting in abnormal expression of the Aurora B and INCENP components of the chromosomal passenger complex. Molecular mechanism studies have found that Src interacts with and phosphorylates INCENP. This then leads to incorrect chromosome arrangement and segregation, resulting in cell division failure. Herein, Src and chromosomal passenger complex co-localize and Src inhibition impedes mitotic progression by inducing multipolar spindle formation. These findings provide novel insights into the molecular basis for using Src inhibitors to treat cancer.


Asunto(s)
Antineoplásicos , Genes src , Mitosis , Proteínas Proto-Oncogénicas pp60(c-src) , Humanos , Proteínas Cromosómicas no Histona/metabolismo , Cromosomas/metabolismo , Citoesqueleto/metabolismo , Genes src/efectos de los fármacos , Mitosis/efectos de los fármacos , Huso Acromático/genética , Huso Acromático/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/antagonistas & inhibidores , Antineoplásicos/farmacología
2.
Cell Death Differ ; 29(9): 1719-1729, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35197565

RESUMEN

Stratification of the epidermis is essential for the barrier function of the skin. However, the molecular mechanisms governing epidermal stratification are not fully understood. Herein, we demonstrate that enkurin domain-containing protein 1 (ENKD1) contributes to epidermal stratification by modulating the cell-division orientation of basal keratinocytes. The epidermis of Enkd1 knockout mice is thinner than that of wild-type mice due to reduced generation of suprabasal cells from basal keratinocytes through asymmetric division. Depletion of ENKD1 impairs proper orientation of the mitotic spindle and delays mitotic progression in cultured cells. Mechanistic investigation further reveals that ENKD1 is a novel microtubule-binding protein that promotes the stability of astral microtubules. Introduction of the microtubule-binding domain of ENKD1 can largely rescue the spindle orientation defects in ENKD1-depleted cells. These findings establish ENKD1 as a critical regulator of astral microtubule stability and spindle orientation that stimulates epidermal stratification in mammalian cells.


Asunto(s)
Microtúbulos , Huso Acromático , Animales , Proteínas de Unión a Calmodulina/metabolismo , Epidermis/metabolismo , Queratinocitos/metabolismo , Mamíferos/metabolismo , Ratones , Ratones Noqueados , Microtúbulos/metabolismo , Mitosis , Proteínas de Plasma Seminal/metabolismo , Piel/metabolismo , Huso Acromático/metabolismo
3.
Cell Tissue Res ; 387(1): 85-93, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34729647

RESUMEN

Human embryonic stem cells (hESCs) are self-renewing and pluripotent cells that originate from the inner cell mass of the blastocyst. Mitosis is fundamental to organism survival and reproduction and is responsible for the equal distribution of duplicated chromosomes into daughter cells. Mitotic dysfunction is associated with a wide variety of human diseases, not least cancer. hESCs have a unique cell cycle distribution, but it is unclear exactly how the mitotic activity of hESCs is related to their proliferation and differentiation. Here, we established a cell line of hESCs stably expressing GFP-α-tubulin and mCherry-H2B by lentiviral infection to analyze and visualize mitosis in detail. During metaphase, the mitotic spindle was smaller and wider and contained a greater proportion of astral microtubules than normal cells. In addition, spindle microtubules were more stable, and chromosome alignment was faster in hESCs than in somatic cells. We also found that the spindle assembly checkpoint was functional in hESCs. These findings thus reveal a specialized mitotic behavior of hESCs.


Asunto(s)
Células Madre Embrionarias Humanas/inmunología , Mitosis/inmunología , Células HeLa , Humanos
4.
Exp Cell Res ; 384(1): 111618, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31505167

RESUMEN

End binding protein 1 (EB1) is a key regulator of microtubule dynamics that orchestrates hierarchical interaction networks at microtubule plus ends to control proper cell division. EB1 activity is known to be regulated by serine/threonine phosphorylation; however, how tyrosine phosphorylation affects EB1 activity remains poorly understood. In this study, we mapped the tyrosine phosphorylation pattern of EB1 in synchronized cells and identified two tyrosine phosphorylation sites (Y217 and Y247) in mitotic cells. Using phospho-deficient (Y/F) and phospho-mimic (Y/D) mutants, we revealed that Y247, but not Y217, is critical for astral microtubule stability. The Y247D mutant contributed to increased spindle angle, indicative of defects in spindle orientation. Time-lapse microscopy revealed that the Y247D mutant significantly delayed mitotic progression by increasing the duration times of prometaphase and metaphase. Structural analysis suggests that Y247 mutants lead to instability of the hydrophobic cavity in the EB homology (EBH) domain, thereby affecting its interactions with p150glued, a protein essential for Gαi/LGN/NuMA complex capture. These findings uncover a crucial role for EB1 phosphorylation in the regulation of mitotic spindle orientation and cell division.


Asunto(s)
Proteínas Asociadas a Microtúbulos/metabolismo , Mitosis/fisiología , Fosforilación/fisiología , Línea Celular Tumoral , Complejo Dinactina/metabolismo , Células HeLa , Humanos , Metafase/fisiología , Microtúbulos/metabolismo , Microtúbulos/fisiología , Unión Proteica/fisiología , Huso Acromático/metabolismo , Huso Acromático/fisiología
5.
Oncotarget ; 8(58): 98233-98241, 2017 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-29228685

RESUMEN

Pancreatic cancer has a poor prognosis due to its rapid rate of metastasis and frequent late-stage diagnosis. An improved understanding of the molecular mechanisms underlying this disease is urgently needed to promote the development of improved diagnostic tools and more effective therapies. Apoptosis signal-regulating kinase 1 (ASK1) has been shown to be overexpressed in pancreatic cancer and to promote the proliferation of pancreatic cancer cells in a kinase activity-dependent manner. However, the molecular mechanisms by which ASK1 promotes cell proliferation remain to be elucidated. In this study, we report that the phosphorylation of end-binding protein 1 (EB1) at threonine 206 (pT206-EB1), which is catalyzed by ASK1, is increased in pancreatic cancer tissues. We further find that the level of pT206-EB1 correlates with that of ASK1 in cancer tissues. Additionally, ASK1 localizes to spindle poles, and knockdown of ASK1 results in the formation of multipolar spindles. Moreover, we show that depletion of ASK1 or disruption of EB1 phosphorylation inhibits spindle microtubule dynamics in pancreatic cancer cells. Collectively, these findings suggest that EB1 phosphorylation mediates the functions of ASK1 in pancreatic cancer development.

6.
Oncotarget ; 8(6): 9858-9867, 2017 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-28039481

RESUMEN

Phosphorylation of end-binding protein 1 (EB1), a key member of microtubule plus end-tracking proteins (+TIPs), by apoptosis signal-regulating kinase 1 (ASK1) has been demonstrated to promote the stability of astral microtubules during mitosis by stimulating the binding of EB1 to microtubule plus ends. However, the roles of other members of the +TIPs family in ASK1/EB1-mediated regulation of astral microtubules are unknown. Herein, we show that ASK1-mediated phosphorylation of EB1 enhances the localization of cytoplasmic linker protein 170 (CLIP-170) and p150glued to the plus ends of astral microtubules. Depletion of ASK1 or expression of phospho-deficient or phospho-mimetic EB1 mutants results in changes in the levels of plus-end localized CLIP-170 or p150glued. Mechanistic studies reveal that EB1 phosphorylation promotes its interactions with CLIP-170 and p150glued, thereby recruiting these +TIPs to microtubules. Structural analysis suggests that serine-40 is the primary phosphorylation site on EB1 that exerts these effects. Together, these findings provide novel insight into the molecular mechanisms that regulate the interactions of EB1 with other +TIPs.


Asunto(s)
Complejo Dinactina/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias del Cuello Uterino/metabolismo , Femenino , Células HeLa , Humanos , MAP Quinasa Quinasa Quinasa 5/genética , MAP Quinasa Quinasa Quinasa 5/metabolismo , Proteínas Asociadas a Microtúbulos/química , Proteínas Asociadas a Microtúbulos/genética , Microtúbulos/genética , Microtúbulos/patología , Mutación , Fosforilación , Unión Proteica , Conformación Proteica , Interferencia de ARN , Transducción de Señal , Relación Estructura-Actividad , Transfección , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/patología
7.
Cell Discov ; 2: 16033, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27721984

RESUMEN

Orientation and positioning of the mitotic spindle are involved in dictating cell division axis and cleavage site, and play important roles in cell fate determination and tissue morphogenesis. However, how spindle movement is controlled to achieve a defined alignment within the dividing cell is not fully understood. Here, we describe an unexpected role for apoptosis signal-regulating kinase 1 (ASK1) in regulating spindle behavior. We find that ASK1 is required for proper mitotic progression and daughter cell adhesion to the substratum. ASK1 interacts with end-binding protein 1 (EB1) and phosphorylates EB1 at serine 40, threonine 154 and threonine 206, enhancing its binding to the plus ends of astral microtubules. Consequently, astral microtubules are stabilized and therefore capable of mediating spindle interaction with the cell cortex, a requirement for spindle movement. These findings reveal a previously undiscovered function of ASK1 in cell division by regulating spindle orientation and positioning, and point to the importance of protein phosphorylation in the regulation of spindle behavior.

8.
Theranostics ; 6(12): 2129-2140, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27698945

RESUMEN

Cell migration, a complex process critical for tumor progression and metastasis, requires a dynamic crosstalk between microtubules (MTs) and focal adhesions (FAs). However, the molecular mechanisms underlying this event remain elusive. Herein we identify the proto-oncogenic protein Src as an important player in the regulation of the MT-FA crosstalk. Src interacts with and phosphorylates end-binding protein 1 (EB1), a member of MT plus end-tracking proteins (+TIPs), both in cells and in vitro. Systematic mutagenesis reveals that tyrosine-247 (Y247) is the primary residue of EB1 phosphorylated by Src. Interestingly, both constitutively activated Src and Y247-phosphorylated EB1 localize to the centrosome and FAs. Src-mediated EB1 phosphorylation diminishes its interactions with other +TIPs, including adenomatous polyposis coli (APC) and mitotic centromere associated kinesin (MCAK). In addition, EB1 phosphorylation at Y247 enhances the rate of MT catastrophe and significantly stimulates cell migration. These findings thus demonstrate that the Src-EB1 axis plays a crucial role in regulating the crosstalk between MTs and FAs to promote cell migration.


Asunto(s)
Movimiento Celular , Adhesiones Focales , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Centrosoma/química , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana , Humanos , Fosforilación , Unión Proteica , Mapeo de Interacción de Proteínas
9.
Oncotarget ; 7(46): 75155-75164, 2016 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-27655673

RESUMEN

Pancreatic cancer has an extremely grim prognosis, with an overall 5-year survival rate less than 5%, as a result of its rapid metastasis and late diagnosis. To combat this disease, it is crucial to better understand the molecular mechanisms that contribute to its pathogenesis. Herein, we report that apoptosis signal-regulating kinase 1 (ASK1) is overexpressed in pancreatic cancer tissues and that its expression correlates with the histological grade of pancreatic cancer. The expression of ASK1 is also elevated in pancreatic cancer cell lines at both protein and mRNA levels. In addition, ASK1 promotes the proliferation and stimulates the tumorigenic capacity of pancreatic cancer cells. These functions of ASK1 are abrogated by pharmacological inhibition of its kinase activity or by introduction of a kinase-dead mutation, suggesting that the kinase activity of ASK1 is required for its role in pancreatic cancer. However, the alteration of ASK1 expression or activity does not significantly affect the migration or invasion of pancreatic cancer cells. Collectively, these findings reveal a critical role for ASK1 in the development of pancreatic cancer and have important implications for the diagnosis and treatment of this malignancy.


Asunto(s)
MAP Quinasa Quinasa Quinasa 5/metabolismo , Proteínas Oncogénicas/metabolismo , Neoplasias Pancreáticas/metabolismo , Animales , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , MAP Quinasa Quinasa Quinasa 5/genética , Ratones , Proteínas Oncogénicas/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Carga Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Theranostics ; 5(7): 656-66, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25897332

RESUMEN

Noscapine is an orally administrable drug used worldwide for cough suppression and has recently been demonstrated to disrupt microtubule dynamics and possess anticancer activity. However, the molecular mechanisms regulating noscapine activity remain poorly defined. Here we demonstrate that cylindromatosis (CYLD), a microtubule-associated tumor suppressor protein, modulates the activity of noscapine both in cell lines and in primary cells of acute lymphoblastic leukemia (ALL). Flow cytometry and immunofluorescence microscopy reveal that CYLD increases the ability of noscapine to induce mitotic arrest and apoptosis. Examination of cellular microtubules as well as in vitro assembled microtubules shows that CYLD enhances the effect of noscapine on microtubule polymerization. Microtubule cosedimentation and fluorescence titration assays further reveal that CYLD interacts with microtubule outer surface and promotes noscapine binding to microtubules. These findings thus demonstrate CYLD as a critical regulator of noscapine activity and have important implications for ALL treatment.


Asunto(s)
Microtúbulos/efectos de los fármacos , Noscapina/farmacología , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Apoptosis , Puntos de Control del Ciclo Celular , Línea Celular Tumoral , Enzima Desubiquitinante CYLD , Humanos , Microtúbulos/metabolismo , Mitosis
11.
DNA Cell Biol ; 34(4): 290-5, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25621430

RESUMEN

Signal transducer and activator of transcription 3 (STAT3) is an important oncogenic transcription factor residing in the cytoplasm in the resting cells. Upon stimulation, STAT3 is activated and translocated to the nucleus to regulate target genes. Although the canonical transcriptional function of STAT3 has been intensively studied, less is known about its cytoplasmic localization. In this study, by immunoprecipitation, microtubule cosedimentation, and immunofluorescence assays, we present the first evidence that cytoplasmic STAT3 interacts with both tubulin and microtubules. By using small-molecule inhibitor approaches, we further demonstrate that the localization of STAT3 on microtubules and its activation are independent of histone deacetylase 6 (HDAC6) activity. In addition, disruption of microtubule dynamics does not alter the activation and nuclear translocation of STAT3 in response to interleukin-6 treatment. These findings reveal that cytoplasmic STAT3 is physically associated with microtubules, whereas its activation and nuclear translocation are independent of microtubule dynamics, implicating that the association of STAT3 with microtubules might be involved in the regulation of noncanonical functions of STAT3 in the cytoplasm.


Asunto(s)
Histona Desacetilasas/metabolismo , Microtúbulos/metabolismo , Factor de Transcripción STAT3/metabolismo , Acetilación , Western Blotting , Núcleo Celular/metabolismo , Células Cultivadas , Citoplasma/efectos de los fármacos , Citoplasma/metabolismo , Técnica del Anticuerpo Fluorescente , Histona Desacetilasa 6 , Humanos , Inmunoprecipitación , Interleucina-6/farmacología , Microtúbulos/efectos de los fármacos , Tubulina (Proteína)/metabolismo
13.
Protein Cell ; 5(6): 469-79, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24748116

RESUMEN

Paclitaxel is a microtubule-targeting agent widely used for the treatment of many solid tumors. However, patients show variable sensitivity to this drug, and effective diagnostic tests predicting drug sensitivity remain to be investigated. Herein, we show that the expression of end-binding protein 1 (EB1), a regulator of microtubule dynamics involved in multiple cellular activities, in breast tumor tissues correlates with the pathological response of tumors to paclitaxel-based chemotherapy. In vitro cell proliferation assays reveal that EB1 stimulates paclitaxel sensitivity in breast cancer cell lines. Our data further demonstrate that EB1 increases the activity of paclitaxel to cause mitotic arrest and apoptosis in cancer cells. In addition, microtubule binding affinity analysis and polymerization/depolymerization assays show that EB1 enhances paclitaxel binding to microtubules and stimulates the ability of paclitaxel to promote microtubule assembly and stabilization. These findings thus reveal EB1 as a critical regulator of paclitaxel sensitivity and have important implications in breast cancer chemotherapy.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/patología , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/metabolismo , Paclitaxel/farmacología , Antineoplásicos Fitogénicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Femenino , Humanos , Células MCF-7 , Proteínas Asociadas a Microtúbulos/antagonistas & inhibidores , Proteínas Asociadas a Microtúbulos/genética , Microtúbulos/química , Paclitaxel/uso terapéutico , Interferencia de ARN , ARN Interferente Pequeño/metabolismo
14.
Cell Cycle ; 13(6): 974-83, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24552808

RESUMEN

Cylindromatosis (CYLD), a deubiquitinase involved in inflammation and tumorigenesis via the modulation of cell signaling, has recently been identified as a critical regulator of microtubule dynamics. CYLD has also been shown to stimulate cell migration and thereby contribute to normal physiological processes. However, it remains elusive how the regulation of microtubule dynamic properties by CYLD is connected to its role in mediating cell migration. In this study, we performed yeast 2-hybrid screening with CYLD as bait and identified 7 CYLD-interacting proteins, including end-binding protein 1 (EB1). The CYLD-EB1 interaction was confirmed both in cells and in vitro, and these 2 proteins colocalized at the plus ends of microtubules. Interestingly, the association of CYLD with EB1 was significantly increased upon the stimulation of cell migration. CYLD coordinated with EB1 to orchestrate tail retraction, centrosome reorientation, and leading-edge microtubule stabilization in migratory cells. In addition, CYLD acted in concert with EB1 to regulate microtubule assembly in vitro, microtubule nucleation at the centrosome, and microtubule growth at the cell periphery. These data provide mechanistic insights into the actions of CYLD in the regulation of microtubule dynamics and cell migration. These findings also support the notion that coordinated actions of microtubule-binding proteins are critical for microtubule-mediated cellular events.


Asunto(s)
Movimiento Celular/fisiología , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/fisiología , Proteínas Supresoras de Tumor/metabolismo , Centrosoma/fisiología , Enzima Desubiquitinante CYLD , Células HeLa , Humanos
15.
Cell Cycle ; 12(7): 1133-41, 2013 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-23470638

RESUMEN

Parkin, an E3 ubiquitin ligase well known for its role in the pathogenesis of juvenile Parkinson disease, has been considered as a candidate tumor suppressor in certain types of cancer. It remains unknown whether parkin is involved in the development of pancreatic cancer, the fourth leading cause of cancer-related deaths worldwide. Herein, we demonstrate the downregulation and copy number loss of the parkin gene in human pancreatic cancer specimens. The expression of parkin negatively correlates with clinicopathological parameters indicating the malignancy of pancreatic cancer. In addition, knockdown of parkin expression promotes the proliferation and tumorigenic properties of pancreatic cancer cells both in vitro and in mice. We further find that parkin deficiency increases the proportion of cells with spindle multipolarity and multinucleation. Parkin-depleted cells also show a significant increase in spindle misorientation. These findings indicate crucial involvement of parkin deficiency in the pathogenesis of pancreatic cancer.


Asunto(s)
Neoplasias Pancreáticas/metabolismo , Huso Acromático/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Proliferación Celular , Regulación hacia Abajo , Dosificación de Gen , Humanos , Cinesinas/antagonistas & inhibidores , Cinesinas/genética , Cinesinas/metabolismo , Ratones , Ratones Desnudos , Neoplasias Pancreáticas/patología , Interferencia de ARN , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Trasplante Heterólogo , Células Tumorales Cultivadas , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores , Ubiquitina-Proteína Ligasas/genética
16.
Acta Pharmacol Sin ; 32(12): 1543-8, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21986572

RESUMEN

AIM: The mitotic kinesin Eg5 plays a critical role in bipolar spindle assembly, and its inhibitors have shown impressive anticancer activity in preclinical studies. This study was undertaken to investigate the effect of dimethylenastron, a specific inhibitor of Eg5, on the migration and invasion of pancreatic cancer cells. METHODS: Human pancreatic cancer cell lines PANC1, EPP85, BxPC3, CFPAC1, and AsPAC1 were used. Eg5 expression was examined using immunofluorescence microscopy. Cell migration and invasion were analyzed with wound healing and transwell assays. Cell proliferation was examined using sulforhodamine B and MTT assays. The binding of dimethylenastron to Eg5 was analyzed with a molecular modeling study, and the ADP release rate was examined with the MANT-ADP reagent. RESULTS: Eg5 expression was 9-16-fold up-regulated in the 5 pancreatic cancer cell lines. Treatment of PANC1 pancreatic cancer cells with dimethylenastron (3 and 10 µmol/L) for 24 h suppressed the migratory ability of the cancer cells in a concentration-dependent manner. The invasion ability of the cancer cells was also reduced by the treatment. However, treatment of PANC1 cells with dimethylenastron (3 and 10 µmol/L) for 24 h had no detectable effect on their proliferation, which was inhibited when the cancer cells were treated with the drug for 72 h. Molecular modeling study showed that dimethylenastron could allosterically inhibit the motor domain ATPase of Eg5 by decreasing the rate of ADP release. CONCLUSION: Dimethylenastron inhibits the migration and invasion of PANC1 pancreatic cancer cells, independent of suppressing the cell proliferation. The findings provide a novel insight into the mechanisms of targeting Eg5 for pancreatic cancer chemotherapy.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Cinesinas/antagonistas & inhibidores , Invasividad Neoplásica/prevención & control , Neoplasias Pancreáticas/patología , Quinazolinas/farmacología , Tionas/farmacología , Regulación Alostérica , Línea Celular Tumoral , Humanos , Neoplasias Pancreáticas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA