Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 114(43): 11500-11505, 2017 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-29073078

RESUMEN

WT p53 is critical for tumor suppression, whereas mutant p53 promotes tumor progression. Nerve injury-induced protein 1 (Ninj1) is a target of p53 and forms a feedback loop with p53 by repressing p53 mRNA translation. Here, we show that loss of Ninj1 increased mutant p53 expression and, subsequently, enhanced cell growth and migration in cells carrying a mutant p53. In contrast, loss of Ninj1 inhibited cell growth and migration in cells carrying a WT p53. To explore the biological significance of Ninj1, we generated a cohort of Ninj1-deficient mice and found that Ninj1+/- mice were prone to systemic inflammation and insulitis, but not to spontaneous tumors. We also found that loss of Ninj1 altered the tumor susceptibility in both mutant p53 and p53-null background. Specifically, in a mutant p53(R270H) background, Ninj1 deficiency shortened the lifespan, altered the tumor spectrum, and increased tumor burden, likely via enhanced expression of mutant p53. In a p53-null background, Ninj1 deficiency significantly increased the incidence of T-lymphoblastic lymphoma. Taken together, our data suggest that depending on p53 genetic status, Ninj1 has two opposing functions in tumorigenesis and that the Ninj1-p53 loop may be targeted to manage inflammatory diseases and cancer.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/metabolismo , Inflamación/genética , Factores de Crecimiento Nervioso/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Alelos , Animales , Moléculas de Adhesión Celular Neuronal/genética , Línea Celular Tumoral , Heterocigoto , Humanos , Inflamación/patología , Longevidad , Ratones , Ratones Transgénicos , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Factores de Crecimiento Nervioso/genética , Proteína p53 Supresora de Tumor/genética
2.
Adv Biol Regul ; 62: 1-10, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27036498

RESUMEN

Inositol 1,4,5-trisphosphate 3-kinase C (ITPKC) is the last identified member of the inositol 1,4,5-trisphosphate 3-kinases family which phosphorylates inositol 1,4,5-trisphosphate into inositol 1,3,4,5-tetrakisphosphate. Although expression and function of the two other family members ITPKA and ITPKB are rather well characterized, similar information is lacking for ITPKC. Here, we first defined the expression of Itpkc mRNA and protein in mouse tissues and cells using in situ hybridization and new antibodies. Surprisingly, we found that cells positive for ITPKC in the studied tissues express either a multicilium (tracheal and bronchial epithelia, brain ependymal cells), microvilli forming a brush border (small and large intestine, and kidney proximal tubule cells) or a flagellum (spermatozoa), suggesting a role for ITPKC either in the development or the function of these specialized cellular structures. Given this surprising expression, we then analyzed ITPKC function in multiciliated tracheal epithelial cells and sperm cells using our Itpkc knock-out mouse model. Unfortunately, no significant difference was observed between control and mutant mice for any of the parameters tested, leaving the exact in vivo function of this third Ins(1,4,5)P3 3-kinase still open.


Asunto(s)
Cilios/enzimología , Células Epiteliales/enzimología , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , ARN Mensajero/genética , Mucosa Respiratoria/enzimología , Secuencia de Aminoácidos , Animales , Encéfalo/enzimología , Cilios/ultraestructura , Células Epiteliales/citología , Expresión Génica , Hibridación in Situ , Inositol 1,4,5-Trifosfato/metabolismo , Fosfatos de Inositol/metabolismo , Intestino Grueso/enzimología , Intestino Delgado/enzimología , Túbulos Renales Proximales/enzimología , Masculino , Ratones , Ratones Noqueados , Fosforilación , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Cultivo Primario de Células , ARN Mensajero/metabolismo , Mucosa Respiratoria/citología , Espermatozoides/enzimología
3.
Genes Dev ; 30(5): 522-34, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26915821

RESUMEN

Poly(C)-binding protein 4 (PCBP4), also called MCG10 and a target of p53, plays a role in the cell cycle and is implicated in lung tumor suppression. Here, we found that PCBP4-deficient mice are prone to lung adenocarcinoma, lymphoma, and kidney tumor and that PCBP4-deficient mouse embryo fibroblasts (MEFs) exhibit enhanced cell proliferation but decreased cellular senescence. We also found that p53 expression is markedly reduced in PCBP4-deficient MEFs and mouse tissues, suggesting that PCBP4 in turn regulates p53 expression. To determine how PCBP4 regulates p53 expression, PCBP4 targets were identified by RNA immunoprecipitation followed by RNA sequencing (RNA-seq). We found that the transcript encoding ZFP871 (zinc finger protein 871; also called ZNF709 in humans) interacts with and is regulated by PCBP4 via mRNA stability. Additionally, we found that ZFP871 physically interacts with p53 and MDM2 proteins. Consistently, ectopic expression of ZFP871 decreases-whereas knockdown of ZFP871 increases-p53 protein stability through a proteasome-dependent degradation pathway. Moreover, loss of ZFP871 reverses the reduction of p53 expression by lack of PCBP4, and thus increased expression of ZFP871 is responsible for decreased expression of p53 in the PCBP4-deficient MEFs and mouse tissues. Interestingly, we found that, like PCBP4, ZFP871 is also regulated by DNA damage and p53. Finally, we showed that knockdown of ZFP871 markedly enhances p53 expression, leading to growth suppression and apoptosis in a p53-dependent manner. Thus, the p53-PCBP4-ZFP871 axis represents a novel feedback loop in the p53 pathway. Together, we hypothesize that PCBP4 is a potential tissue-specific tumor suppressor and that ZFP871 is part of MDM2 and possibly other ubiquitin E3 ligases that target p53 for degradation.


Asunto(s)
Proteínas Portadoras/genética , Regulación Neoplásica de la Expresión Génica , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/fisiopatología , Adenocarcinoma del Pulmón , Animales , Proliferación Celular/genética , Senescencia Celular/genética , Proteínas de Unión al ADN , Técnicas de Silenciamiento del Gen , Estimación de Kaplan-Meier , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/fisiopatología , Ratones , Unión Proteica , Estabilidad Proteica , Proteolisis
4.
FEBS J ; 282(13): 2553-69, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25892505

RESUMEN

Four inositol phosphate kinases catalyze phosphorylation of the second messenger inositol 1,4,5-trisphosphate [Ins(1,4,5)P3 ] to inositol 1,3,4,5-tetrakisphosphate [Ins(1,3,4,5)P4 ]: these enzymes comprise three isoenzymes of inositol 1,4,5-trisphosphate 3-kinase (Itpk), referred to as Itpka, Itpkb and Itpkc, and the inositol polyphosphate multikinase (IPMK). The four enzymes that act on Ins(1,4,5)P3 are all expressed in rat pheochromocytoma PC12 cells, a model that is used to study neurite outgrowth induced by nerve growth factor (NGF). We compared the effect of over-expression of the four GFP-tagged kinases on NGF-induced neurite outgrowth. Our data show that over-expression of the Itpka and Itpkb isoforms inhibits NGF-induced neurite outgrowth, but over-expression of Itpkc and IPMK does not. Surprisingly, over-expression of the N-terminal F-actin binding domain of Itpka, which lacks catalytic activity, was as effective at inhibiting neurite outgrowth as the full-length enzyme. Neurite length was also significantly decreased in cells over-expressing Itpka and Itpkb but not Itpkc or IPMK. This result did not depend on the over-expression level of any of the kinases. PC12 cells over-expressing GFP-tagged kinase-dead mutants Itpka/b have shorter neurites than GFP control cells. The decrease in neurite length was never as pronounced as observed with wild-type GFP-tagged Itpka/b. Finally, the percentage of neurite-bearing cells was increased in cells over-expressing the membranous type I Ins(1,4,5)P3 5-phosphatase. We conclude that Itpka and Itpkb inhibit neurite outgrowth through both F-actin binding and localized Ins(1,4,5)P3 3-kinase activity. Itpkc and IPMK do not influence neurite outgrowth or neurite length in this model.


Asunto(s)
Isoenzimas/fisiología , Factor de Crecimiento Nervioso/farmacología , Neuritas/fisiología , Fosfotransferasas (Aceptor de Grupo Alcohol)/fisiología , Actinas/química , Animales , Proteínas Fluorescentes Verdes/metabolismo , Células PC12 , Ratas
5.
Adv Biol Regul ; 57: 153-61, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25294679

RESUMEN

RASA3 (or GTPase Activating Protein III, R-Ras GTPase-activating protein, GAP1(IP4BP)) is a GTPase activating protein of the GAP1 subfamily which targets Ras and Rap1. RASA3 was originally purified from pig platelet membranes through its intrinsic ability to bind inositol 1,3,4,5-tetrakisphosphate (I(1,3,4,5)P4) with high affinity, hence its first name GAP1(IP4BP) (for GAP1 subfamily member which binds I(1,3,4,5)P4). RASA3 was thus the first I(1,3,4,5)P4 receptor identified and cloned. The in vitro and in vivo functions of RASA3 remained somewhat elusive for a long time. However, recently, using genetically-modified mice and cells derived from these mice, the function of RASA3 during megakaryopoiesis, megakaryocyte adhesion and migration as well as integrin signaling has been reported. The goal of this review is thus to summarize and comment recent and less recent data in the literature on RASA3, in particular on the in vivo function of this specific GAP1 subfamily member.


Asunto(s)
Movimiento Celular/fisiología , Proteínas Activadoras de GTPasa , Receptores Citoplasmáticos y Nucleares , Transducción de Señal/fisiología , Trombopoyesis/fisiología , Animales , Adhesión Celular/fisiología , Proteínas Activadoras de GTPasa/genética , Proteínas Activadoras de GTPasa/metabolismo , Humanos , Fosfatos de Inositol , Ratones , Ratones Transgénicos , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo
6.
Nucleic Acids Res ; 39(1): 213-24, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20817677

RESUMEN

RNA-binding proteins (RBPs) play a major role in many post-transcriptional processes, including mRNA stability, alternative splicing and translation. PCBP4, also called MCG10, is an RBP belonging to the poly(C)-binding protein family and a target of p53 tumor suppressor. Ectopic expression of PCBP4 induces cell-cycle arrest in G2 and apoptosis. To identify RNA targets regulated by PCBP4 and further decipher its function, we generated multiple cell lines in which PCBP4 is either inducibly over-expressed or knocked down. We found that PCBP4 expression decreases cyclin-dependent kinase inhibitor p21 induction in response to DNA damage. We also provided evidence that PCBP4 regulates p21 expression independently of p53. In addition, we showed that a deficiency in PCBP4 enhances p21 induction upon DNA damage. To validate PCBP4 regulation of p21, we made PCBP4-deficient mice and showed that p21 expression is markedly increased in PCBP4-deficient primary mouse embryo fibroblasts compared to that in wild-type counterparts. Finally, we uncovered that PCBP4 binds to the 3'-UTR of p21 transcript in vitro and in vivo to regulate p21 mRNA stability. Taken together, we revealed that PCBP4 regulates both basal and stress-induced p21 expression through binding p21 3'-UTR and modulating p21 mRNA stability.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Estabilidad del ARN , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/fisiología , Regiones no Traducidas 3' , Empalme Alternativo , Animales , Línea Celular Tumoral , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/biosíntesis , Daño del ADN , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Ratones Noqueados , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
7.
Mol Cell Pharmacol ; 2(3): 111-116, 2010 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-20686680

RESUMEN

G protein-coupled receptors (GPRs) constitute one of the largest families of membrane proteins encoded by the human genome. Upon binding to various ligands, these seven-transmembrane receptors play an essential role in many physiological processes, including neurotransmission, immunity, inflammation, regulation of mood and behavior. In view of their important functions, aberrant expression and activity of GPRs have been implicated in a wide spectrum of diseases, including tumorigenesis. GPR87, a cell surface GPR related to the LPA receptor family, is overexpressed in diverse carcinomas and plays an essential role in tumor cell survival. In our recent work, we uncovered that GPR87 expression is regulated by the tumor suppressor p53 and by DNA damage in a p53-dependent manner. Moreover, we found that a lack of GPR87 triggers an increase in p53, concomitant with a decrease in Akt, which results in the sensitization of tumor cells to DNA damage-induced apoptosis and growth suppression. Altogether, we uncovered an essential function for GPR87 in p53-dependent cell survival in response to stress signals. Due to their unique structure, localization and ligand binding ability, GPRs have been extensively used for drug development and are the most common targets of commercial drugs. Although studies are required to determine GPR87 natural ligand(s) and signaling pathways, GPR87 is undoubtedly a very promising novel target for cancer prevention and treatment.

8.
Cancer Res ; 68(16): 6789-96, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-18701504

RESUMEN

Overexpression of mutant p53 is a common theme in human tumors, suggesting a tumor-promoting gain-of-function for mutant p53. To elucidate whether and how mutant p53 acquires its gain-of-function, mutant p53 is inducibly knocked down in the SW480 colon cancer cell line, which contains mutant p53(R273H/P309S), and the MIA PaCa-2 pancreatic cancer cell line, which contains mutant p53(R248W). We found that knockdown of mutant p53 markedly inhibits cell proliferation. In addition, knockdown of mutant p53 sensitizes tumor cells to growth suppression by various chemotherapeutic drugs. To determine whether a gene involved in cell growth and survival is regulated by mutant p53, gene expression profiling analysis was performed and showed that the expression level of Id2, a member of the inhibitor of differentiation (Id) family, was markedly increased upon knockdown of mutant p53. To confirm this, Northern blot analysis was performed and showed that the expression level of Id2 was regulated by various mutant p53s in multiple cell lines. In addition, we found that the Id2 promoter is responsive to mutant but not wild-type p53, and mutant p53 binds to the Id2 promoter. Consistent with these observations, expression of endogenous Id2 was found to be inhibited by exogenous mutant p53 in p53-null HCT116 cells. Finally, we showed that knockdown of Id2 can restore the proliferative potential of tumor cells inhibited by withdrawal of mutant p53. Together, these findings suggest that one mechanism by which mutant p53 acquires its gain-of-function is through the inhibition of Id2 expression.


Asunto(s)
Proliferación Celular , Proteína 2 Inhibidora de la Diferenciación/antagonistas & inhibidores , Proteína 2 Inhibidora de la Diferenciación/fisiología , Mutación/genética , Proteína p53 Supresora de Tumor/genética , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Northern Blotting , Inmunoprecipitación de Cromatina , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Ensayo de Unidades Formadoras de Colonias , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Luciferasas/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Regiones Promotoras Genéticas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/metabolismo
9.
J Biol Chem ; 282(21): 15471-5, 2007 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-17409384

RESUMEN

The lysine-specific demethylase 1 (LSD1), a component of several histone deacetylase complexes, plays an important role in chromatin remodeling and transcriptional regulation. Here, we generated multiple cell lines in which LSD1 is inducibly expressed or knocked down and found that LSD1 is required for cell proliferation. In addition, we found that deficiency in LSD1 leads to a partial cell cycle arrest in G(2)/M and sensitizes cells to growth suppression induced by DNA damage or MDM2 inhibition in a p53-dependent manner. We also showed that LSD1 deficiency delays p53 stabilization induced by DNA damage, leading to a delayed induction of p21 and MDM2. Finally, we performed a microarray study and identified several novel LSD1 target genes, including S100A8, which encodes a calcium-binding protein, and DEK, a proto-oncogene. Taken together, we uncovered that LSD1 has a pro-oncogenic function by modulating pro-survival gene expression and p53 transcriptional activity.


Asunto(s)
División Celular/fisiología , Ensamble y Desensamble de Cromatina/fisiología , Fase G2/fisiología , Oxidorreductasas N-Desmetilantes/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Calgranulina A/biosíntesis , Calgranulina A/genética , Línea Celular Tumoral , Proteínas Cromosómicas no Histona/biosíntesis , Proteínas Cromosómicas no Histona/genética , Daño del ADN/genética , Perfilación de la Expresión Génica , Histona Demetilasas , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Oncogénicas/biosíntesis , Proteínas Oncogénicas/genética , Oxidorreductasas N-Desmetilantes/deficiencia , Proteínas de Unión a Poli-ADP-Ribosa , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-mdm2/genética , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Proteína p53 Supresora de Tumor/genética
10.
Cancer Res ; 66(12): 6271-9, 2006 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-16778203

RESUMEN

The epithelial cell transforming sequence 2 (ECT2), a member of the Dbl family of guanine nucleotide exchange factor for Rho GTPases, is required for cytokinesis. The tumor suppressor p53 plays a crucial role in coordinating cellular processes, such as cell cycle arrest and apoptosis, in response to stress signals. Here, we showed that ECT2 is negatively regulated by wild-type p53 but not tumor-derived mutant p53 or other p53 family members. In addition, ECT2 is down-regulated in multiple cell lines by DNA damage agents and Nutlin-3, an MDM2 antagonist, in a p53-dependent manner. We also showed that the activity of the ECT2 promoter is repressed by wild-type p53, and to a lesser extent, by p21. In addition, the second activation domain in p53 is necessary for the efficient repression of ECT2. Importantly, we found that the ECT2 gene is bound by p53 in vivo in response to DNA damage and Nutlin-3 treatment. Furthermore, we provided evidence that inhibition of protein methyltransferases, especially arginine methyltransferases, relieve the repression of ECT2 induced by DNA damage or Nutlin-3 in a p53-dependent manner. Finally, we generated multiple cell lines in which ECT2 is inducibly knocked down and found that ECT2 knockdown triggers cell cycle arrest in G1. Taken together, we uncovered a novel function for ECT2 and provided a novel mechanism by which p53 represses gene expression via protein methyltransferases.


Asunto(s)
Fase G1/fisiología , Metiltransferasas/metabolismo , Proteínas Proto-Oncogénicas/fisiología , Fase S/fisiología , Proteína p53 Supresora de Tumor/fisiología , Proteínas de Unión al GTP rho/genética , Línea Celular Tumoral , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Daño del ADN , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Humanos , Imidazoles/farmacología , Péptidos y Proteínas de Señalización Intracelular , Metiltransferasas/antagonistas & inhibidores , Piperazinas/farmacología , Regiones Promotoras Genéticas , Proteína-Arginina N-Metiltransferasas , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteínas de Unión al GTP rho/metabolismo
11.
Cancer Biol Ther ; 4(11): 1178-85, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16294021

RESUMEN

The p53 tumor suppressor is a modular transcription factor that determines cellular outcome (cell cycle arrest and DNA repair vs. apoptosis) in response to stress signals. The two p53 homologues, p63 and p73 play an important role in development but also act as tumor suppressors. The p53 family members are highly homologous in the activation domain (AD), the DNA-binding domain (DBD) and the tetramerization domain (TD) but differ in the C-terminus. Indeed, the p53 C-terminus contains a basic domain (BD) whereas p63/p73 have a sterile alpha motif (SAM) domain and an inhibitory domain (ID). In addition to the full-length proteins, the p53 family genes produce multiple isoforms truncated at the NH2- and/or C-terminus. Importantly, every functional domain is a determinant in the transactivation of specific target genes by the p53 family members. Distinct post-translational modifications and interactions with cofactors further modulate the transcriptional activity of the p53 family members in response to particular stress signals. Therefore, divergence in the composition of the p53 family proteins is responsible for the diversity of p53 family functions.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Genes p53 , Proteína p53 Supresora de Tumor/química , Proteína p53 Supresora de Tumor/fisiología , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Humanos , Señales de Localización Nuclear , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Estructura Terciaria de Proteína , Relación Estructura-Actividad , Activación Transcripcional , Proteína p53 Supresora de Tumor/genética
12.
Atherosclerosis ; 160(1): 59-67, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11755923

RESUMEN

Primary human endothelial cells have a finite life span in vitro. After 3-4 passages, they tend to de-differentiate and eventually reach senescence. This limits their use in studies of endothelial cell function. To overcome this, we have developed human saphenous vein endothelial cell lines (HSVEC lines). Two cell lines were produced by infection with pZipSVtsA58-U19 which encodes the simian virus 40 large T-antigen, and one cell line was obtained by transfection with pLXSN16E6E7, which encodes the human papillomavirus type 16 E6 and E7 genes. Two of the three HSVEC lines exhibited an extended life span in vitro and retained characteristic endothelial "cobblestone" morphology. These cell lines expressed the known endothelial markers CD31 and vascular endothelial cadherin, and were able to bind Ulex europaeus lectin I, but they did not retain the expression of von Willebrand factor. Furthermore, one cell line was able to functionally up-regulate the expression of intercellular adhesion molecule-1 in response to stimulation with tumor necrosis factor alpha and was also able to incorporate acetylated low-density lipoprotein. Our results suggest that this latter HSVEC line will provide a useful resource to investigate selected responses of the vascular endothelium to physiological and pathological situations.


Asunto(s)
Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Vena Safena/citología , Vena Safena/metabolismo , Anticuerpos/inmunología , Antígenos CD , Antígenos Virales de Tumores/biosíntesis , Antígenos Virales de Tumores/inmunología , Cadherinas , División Celular/efectos de los fármacos , Línea Celular , Relación Dosis-Respuesta a Droga , Humanos , Inmunohistoquímica , Molécula 1 de Adhesión Intercelular/biosíntesis , Molécula 1 de Adhesión Intercelular/efectos de los fármacos , Interleucina-1/farmacología , Lipoproteínas LDL/farmacología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta , Retroviridae/inmunología , Infecciones por Retroviridae/inmunología , Factores de Tiempo , Factor de Necrosis Tumoral alfa/farmacología , Regulación hacia Arriba/efectos de los fármacos , Factor de von Willebrand/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...