Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 163
Filtrar
1.
Sci Rep ; 11(1): 17759, 2021 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-34493746

RESUMEN

Inducing cardiac myocytes to proliferate is considered a potential therapy to target heart disease, however, modulating cardiac myocyte proliferation has proven to be a technical challenge. The Hippo pathway is a kinase signaling cascade that regulates cell proliferation during the growth of the heart. Inhibition of the Hippo pathway increases the activation of the transcription factors YAP/TAZ, which translocate to the nucleus and upregulate transcription of pro-proliferative genes. The Hippo pathway regulates the proliferation of cancer cells, pluripotent stem cells, and epithelial cells through a cell-cell contact-dependent manner, however, it is unclear if cell density-dependent cell proliferation is a consistent feature in cardiac myocytes. Here, we used cultured human iPSC-derived cardiac myocytes (hiCMs) as a model system to investigate this concept. hiCMs have a comparable transcriptome to the immature cardiac myocytes that proliferate during heart development in vivo. Our data indicate that a dense syncytium of hiCMs can regain cell cycle activity and YAP expression and activity when plated sparsely or when density is reduced through wounding. We found that combining two small molecules, XMU-MP-1 and S1P, increased YAP activity and further enhanced proliferation of low-density hiCMs. Importantly, these compounds had no effect on hiCMs within a dense syncytium. These data add to a growing body of literature that link Hippo pathway regulation with cardiac myocyte proliferation and demonstrate that regulation is restricted to cells with reduced contact inhibition.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Miocitos Cardíacos/citología , Proteínas Serina-Treonina Quinasas/fisiología , Transducción de Señal/fisiología , Proteínas Adaptadoras Transductoras de Señales/fisiología , Secuencia de Bases , Recuento de Células , Ciclo Celular/efectos de los fármacos , Diferenciación Celular , División Celular/efectos de los fármacos , Células Cultivadas , Inhibición de Contacto/efectos de los fármacos , Vía de Señalización Hippo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Lisofosfolípidos/farmacología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Polimorfismo de Nucleótido Simple , ARN/biosíntesis , ARN/genética , Transducción de Señal/efectos de los fármacos , Esfingosina/análogos & derivados , Esfingosina/farmacología , Sulfonamidas/farmacología , Factores de Transcripción/fisiología , Proteínas Señalizadoras YAP
2.
Biochem Biophys Res Commun ; 546: 65-73, 2021 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-33571906

RESUMEN

The occurrence and development of tumors cannot be separated from the influence of differentiation at different stages and levels. Our study found that E-cadherin was significantly increased in cell model induced by sodium butyrate and cell density, while METTL3, METTL16 and WTAP were decreased during the differentiation of cells. In the clinicopathological tissues, E-cadherin was low expressed in poorly differentiated tumor tissues and above three regulators were highly expressed in poorly differentiated tissues. At the levels of clinicopathological differentiation, tissue differentiation and cell differentiation, the result indicated that the poor prognosis of colorectal cancer (CRC) may be closely related to high expression of total m6A level and high expression of METTL3, METTL16 and WTAP.


Asunto(s)
Adenosina/análogos & derivados , Diferenciación Celular , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Adenosina/metabolismo , Ácido Butírico/farmacología , Proteínas de Ciclo Celular , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Neoplasias Colorrectales/genética , Inhibición de Contacto/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Metiltransferasas , Persona de Mediana Edad , Biosíntesis de Proteínas/efectos de los fármacos , Factores de Empalme de ARN , Transcripción Genética/efectos de los fármacos
3.
PLoS Genet ; 14(11): e1007788, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30418965

RESUMEN

Estrogen therapy increases the risk of ovarian cancer and exogenous estradiol accelerates the onset of ovarian cancer in mouse models. Both in vivo and in vitro, ovarian surface epithelial (OSE) cells exposed to estradiol develop a subpopulation that loses cell polarity, contact inhibition, and forms multi-layered foci of dysplastic cells with increased susceptibility to transformation. Here, we use single-cell RNA-sequencing to characterize this dysplastic subpopulation and identify the transcriptional dynamics involved in its emergence. Estradiol-treated cells were characterized by up-regulation of genes associated with proliferation, metabolism, and survival pathways. Pseudotemporal ordering revealed that OSE cells occupy a largely linear phenotypic spectrum that, in estradiol-treated cells, diverges towards cell state consistent with the dysplastic population. This divergence is characterized by the activation of various cancer-associated pathways including an increase in Greb1 which was validated in fallopian tube epithelium and human ovarian cancers. Taken together, this work reveals possible mechanisms by which estradiol increases epithelial cell susceptibility to tumour initiation.


Asunto(s)
Estradiol/efectos adversos , Ovario/efectos de los fármacos , Ovario/metabolismo , Animales , Polaridad Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Células Cultivadas , Inhibición de Contacto/efectos de los fármacos , Epitelio/efectos de los fármacos , Epitelio/metabolismo , Epitelio/patología , Femenino , Humanos , Proteínas de la Membrana , Ratones , Ovario/patología , Fenotipo , Proteínas/genética , Proteínas/metabolismo , Análisis de Secuencia de ARN , Transducción de Señal/efectos de los fármacos , Análisis de la Célula Individual
4.
Development ; 144(13): 2456-2468, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28526750

RESUMEN

A fundamental property of neural crest (NC) migration is contact inhibition of locomotion (CIL), a process by which cells change their direction of migration upon cell contact. CIL has been proven to be essential for NC migration in amphibians and zebrafish by controlling cell polarity in a cell contact-dependent manner. Cell contact during CIL requires the participation of the cell adhesion molecule N-cadherin, which starts to be expressed by NC cells as a consequence of the switch between E- and N-cadherins during epithelial-to-mesenchymal transition (EMT). However, the mechanism that controls the upregulation of N-cadherin remains unknown. Here, we show that platelet-derived growth factor receptor alpha (PDGFRα) and its ligand platelet-derived growth factor A (PDGF-A) are co-expressed in migrating cranial NC. Inhibition of PDGF-A/PDGFRα blocks NC migration by inhibiting N-cadherin and, consequently, impairing CIL. Moreover, we identify phosphatidylinositol-3-kinase (PI3K)/AKT as a downstream effector of the PDGFRα cellular response during CIL. Our results lead us to propose PDGF-A/PDGFRα signalling as a tissue-autonomous regulator of CIL by controlling N-cadherin upregulation during EMT. Finally, we show that once NC cells have undergone EMT, the same PDGF-A/PDGFRα works as an NC chemoattractant, guiding their directional migration.


Asunto(s)
Cadherinas/metabolismo , Movimiento Celular , Inhibición de Contacto , Locomoción , Cresta Neural/citología , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Xenopus laevis/metabolismo , Animales , Movimiento Celular/efectos de los fármacos , Quimiotaxis/efectos de los fármacos , Inhibición de Contacto/efectos de los fármacos , Locomoción/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología
5.
Hum Mol Genet ; 25(19): 4201-4210, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27516388

RESUMEN

Inactivating mutations of the TSC1/TSC2 complex (TSC1/2) cause tuberous sclerosis (TSC), a hereditary syndrome with neurological symptoms and benign hamartoma tumours in the brain. Since TSC effectors are largely unknown in the human brain, TSC patient cortical tubers were used to uncover hyperphosphorylation unique to TSC primary astrocytes, the cell type affected in the brain. We found abnormal hyperphosphorylation of catenin delta-1 S268, which was reversible by mTOR-specific inhibitors. In contrast, in three metastatic astrocytoma cell lines, S268 was under phosphorylated, suggesting S268 phosphorylation controls metastasis. TSC astrocytes appeared epithelial (i.e. tightly adherent, less motile, and epithelial (E)-cadherin positive), whereas wild-type astrocytes were mesenchymal (i.e. E-cadherin negative and highly motile). Despite their epithelial phenotype, TSC astrocytes outgrew contact inhibition, and monolayers sporadically generated tuberous foci, a phenotype blocked by the mTOR inhibitor, Torin1. Also, mTOR-regulated phosphokinase C epsilon (PKCe) activity induced phosphorylation of catenin delta-1 S268, which in turn mediated cell-cell adhesion in astrocytes. The mTOR-dependent, epithelial phenotype of TSC astrocytes suggests TSC1/2 and mTOR tune the phosphorylation level of catenin delta-1 by controlling PKCe activity, thereby regulating the mesenchymal-epithelial-transition (MET). Thus, some forms of TSC could be treated with PKCe inhibitors, while metastasis of astrocytomas might be blocked by PKCe stimulators.


Asunto(s)
Cateninas/genética , Hamartoma/genética , Proteína Quinasa C-epsilon/genética , Serina-Treonina Quinasas TOR/genética , Proteínas Supresoras de Tumor/genética , Astrocitos/efectos de los fármacos , Astrocitos/patología , Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral , Inhibición de Contacto/efectos de los fármacos , Transición Epitelial-Mesenquimal/genética , Hamartoma/patología , Humanos , Naftiridinas/administración & dosificación , Metástasis de la Neoplasia , Fosforilación/efectos de los fármacos , Proteína 1 del Complejo de la Esclerosis Tuberosa , Proteína 2 del Complejo de la Esclerosis Tuberosa , Catenina delta
6.
Sci Rep ; 5: 17812, 2015 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-26640042

RESUMEN

High density lipoproteins (HDLs) play a crucial role in removing excess cholesterol from peripheral tissues. Although their concentration is lower during conditions of high cell growth rate (cancer and infections), their involvement during cell proliferation is not known. To this aim, we investigated the replicative cycles in synchronised Swiss 3T3 fibroblasts in different experimental conditions: i) contact-inhibited fibroblasts re-entering cell cycle after dilution; ii) scratch-wound assay; iii) serum-deprived cells induced to re-enter G1 by FCS, HDL or PDGF. Analyses were performed during each cell cycle up to quiescence. Cholesterol synthesis increased remarkably during the replicative cycles, decreasing only after cells reached confluence. In contrast, cholesteryl ester (CE) synthesis and content were high at 24 h after dilution and then decreased steeply in the successive cycles. Flow cytometry analysis of DiO-HDL, as well as radiolabeled HDL pulse, demonstrated a significant uptake of CE-HDL in 24 h. DiI-HDL uptake, lipid droplets (LDs) and SR-BI immunostaining and expression followed the same trend. Addition of HDL or PDGF partially restore the proliferation rate and significantly increase SR-BI and pAKT expression in serum-deprived cells. In conclusion, cell transition from G0 to G1/S requires CE-HDL uptake, leading to CE-HDL/SR-BI pathway activation and CEs increase into LDs.


Asunto(s)
Fibroblastos/citología , Fase G1 , Lipoproteínas HDL/metabolismo , Fase de Descanso del Ciclo Celular , Animales , Radioisótopos de Carbono , Proliferación Celular/efectos de los fármacos , Colesterol/metabolismo , Ésteres del Colesterol/metabolismo , Inhibición de Contacto/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Fibroblastos/metabolismo , Citometría de Flujo , Fluorescencia , Ratones , Células 3T3 NIH , Fosforilación/efectos de los fármacos , Factor de Crecimiento Derivado de Plaquetas/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores Depuradores/metabolismo , Triglicéridos/metabolismo
7.
Toxicology ; 333: 37-44, 2015 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-25843524

RESUMEN

Inhibition of apoptosis by the ligands of the aryl hydrocarbon receptor (AhR) has been proposed to play a role in their tumor promoting effects on liver parenchymal cells. However, little is presently known about the impact of toxic AhR ligands, such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) on apoptosis in other liver cell types, such as in liver epithelial/progenitor cells. In the present study, we focused on the effects of TCDD on apoptosis regulation in a model of liver progenitor cells, rat WB-F344 cell line, during the TCDD-elicited release from contact inhibition. The stimulation of cell proliferation in this cell line was associated with deregulated expression of a number of genes known to be under transcriptional control of the Hippo signaling pathway, a principal regulatory pathway involved in contact inhibition of cell proliferation. Interestingly, we found that mRNA and protein levels of survivin, a known Hippo target, which plays a role both in cell division and inhibition of apoptosis, were significantly up-regulated in rat liver epithelial cell model, as well as in undifferentiated human liver HepaRG cells. Using the short interfering RNA-mediated knockdown, we confirmed that survivin plays a central role in cell division of WB-F344 cells. When evaluating the effects of TCDD on apoptosis induction by camptothecin, a genotoxic topoisomerase I inhibitor, we observed that the pre-treatment of WB-F344 cells with TCDD increased number of cells with apoptotic nuclear morphology, and it potentiated cleavage of both caspase-3 and poly(ADP-ribose) polymerase I. This indicated that despite the observed up-regulation of survivin, apoptosis induced by the genotoxin was potentiated in the model of rat liver progenitor cells. The present results indicate that, unlike in hepatocytes, AhR agonists may not prevent induction of apoptosis elicited by DNA-damaging agents in a model of rat liver progenitor cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/agonistas , Camptotecina/toxicidad , Inhibición de Contacto/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Hígado/efectos de los fármacos , Proteínas Asociadas a Microtúbulos/metabolismo , Dibenzodioxinas Policloradas/toxicidad , Receptores de Hidrocarburo de Aril/agonistas , Inhibidores de Topoisomerasa I/toxicidad , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Caspasa 3/metabolismo , Línea Celular , Células Epiteliales/metabolismo , Células Epiteliales/patología , Humanos , Proteínas Inhibidoras de la Apoptosis/genética , Proteínas Inhibidoras de la Apoptosis/metabolismo , Hígado/metabolismo , Hígado/patología , Proteínas Asociadas a Microtúbulos/genética , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/metabolismo , Interferencia de ARN , Ratas Endogámicas F344 , Receptores de Hidrocarburo de Aril/metabolismo , Transducción de Señal/efectos de los fármacos , Survivin , Factores de Tiempo , Transcripción Genética/efectos de los fármacos , Transfección , Regulación hacia Arriba
8.
Cell Cycle ; 14(8): 1197-206, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25695744

RESUMEN

Human corneal endothelial cells (HCECs) responsible for corneal transparency have limited proliferative capacity in vivo because of "contact-inhibition." This feature has hampered the ability to engineer HCECs for transplantation. Previously we have reported an in vitro model of HCECs in which contact inhibition was re-established at Day 21, even though cell junction and cell matrix interaction were not perturbed during isolation. Herein, we observe that such HCEC monolayers continue to expand and retain a normal phenotype for 2 more weeks if cultured in a leukemia inhibitory factor (LIF)-containing serum-free medium. Such expansion is accompanied initially by upregulation of Cyclin E2 colocalized with nuclear translocation of phosphorylated retinoblastoma tumor suppressor (p-Rb) at Day 21 followed by a delay in contact inhibition through activation of LIF-Janus kinase1 (JAK1)-signal transducer and activator of transcription 3 (STAT3) signaling at Day 35. The LIF-JAK1-STAT3 signaling is coupled with upregulation of E2F2 colocalized with nuclear p-Rb and with concomitant downregulation of p16(INK4a), of which upregulation is linked to senescence. Hence, activation of LIF-JAK1-STAT3 signaling to delay contact inhibition can be used as another strategy to facilitate engineering of HCEC grafts to solve the unmet global shortage of corneal grafts.


Asunto(s)
Córnea/citología , Células Endoteliales/citología , Janus Quinasa 1/metabolismo , Factor Inhibidor de Leucemia/farmacología , Factor de Transcripción STAT3/metabolismo , Adolescente , Adulto , Anciano , Núcleo Celular/metabolismo , Células Cultivadas , Inhibición de Contacto/efectos de los fármacos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Factor de Transcripción E2F2/metabolismo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Factor 2 de Crecimiento de Fibroblastos/farmacología , Humanos , Janus Quinasa 1/antagonistas & inhibidores , Janus Quinasa 1/genética , Factor Inhibidor de Leucemia/metabolismo , Persona de Mediana Edad , ARN Interferente Pequeño/metabolismo , Proteína de Retinoblastoma/metabolismo , Factor de Transcripción STAT3/antagonistas & inhibidores , Factor de Transcripción STAT3/genética , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Adulto Joven
9.
Proc Natl Acad Sci U S A ; 111(48): 17188-93, 2014 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-25404301

RESUMEN

Normal human and murine fibroblasts can inhibit proliferation of tumor cells when cocultured in vitro. The inhibitory capacity varies depending on the donor and the site of origin of the fibroblast. We showed previously that effective inhibition requires formation of a morphologically intact fibroblast monolayer before seeding of the tumor cells. Here we show that inhibition is extended to motility of tumor cells and we dissect the factors responsible for these inhibitory functions. We find that inhibition is due to two different sets of molecules: (i) the extracellular matrix (ECM) and other surface proteins of the fibroblasts, which are responsible for contact-dependent inhibition of tumor cell proliferation; and (ii) soluble factors secreted by fibroblasts when confronted with tumor cells (confronted conditioned media, CCM) contribute to inhibition of tumor cell proliferation and motility. However, conditioned media (CM) obtained from fibroblasts alone (nonconfronted conditioned media, NCM) did not inhibit tumor cell proliferation and motility. In addition, quantitative PCR (Q-PCR) data show up-regulation of proinflammatory genes. Moreover, comparison of CCM and NCM with an antibody array for 507 different soluble human proteins revealed differential expression of growth differentiation factor 15, dickkopf-related protein 1, endothelial-monocyte-activating polypeptide II, ectodysplasin A2, Galectin-3, chemokine (C-X-C motif) ligand 2, Nidogen1, urokinase, and matrix metalloproteinase 3.


Asunto(s)
Movimiento Celular/fisiología , Proliferación Celular , Inhibición de Contacto/fisiología , Fibroblastos/citología , Animales , Línea Celular , Línea Celular Tumoral , Células Cultivadas , Técnicas de Cocultivo , Inhibición de Contacto/efectos de los fármacos , Medios de Cultivo Condicionados/química , Medios de Cultivo Condicionados/metabolismo , Medios de Cultivo Condicionados/farmacología , Matriz Extracelular/metabolismo , Matriz Extracelular/fisiología , Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ratones , Microscopía Fluorescente , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína Fluorescente Roja
11.
Cell Mol Biol Lett ; 19(1): 77-97, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24470116

RESUMEN

Cellular adhesion to the underlying substratum is regulated through numerous signaling pathways. It has been suggested that insulin receptor substrate 1 (IRS-1) is involved in some of these pathways, via association with and activation of transmembrane integrins. Calreticulin, as an important endoplasmic reticulum-resident, calcium-binding protein with a chaperone function, plays an obvious role in proteomic expression. Our previous work showed that calreticulin mediates cell adhesion not only by affecting protein expression but also by affecting the state of regulatory protein phosphorylation, such as that of c-src. Here, we demonstrate that calreticulin affects the abundance of IRS-1 such that the absence of calreticulin is paralleled by a decrease in IRS-1 levels and the unregulated overexpression of calreticulin is accompanied by an increase in IRS-1 levels. These changes in the abundance of calreticulin and IRS-1 are accompanied by changes in cell-substratum adhesiveness and phosphorylation, such that increases in the expression of calreticulin and IRS-1 are paralleled by an increase in focal contact-based cell-substratum adhesiveness, and a decrease in the expression of these proteins brings about a decrease in cell-substratum adhesiveness. Wild type and calreticulin-null mouse embryonic fibroblasts (MEFs) were cultured and the IRS-1 isoform profile was assessed. Differences in morphology and motility were also quantified. While no substantial differences in the speed of locomotion were found, the directionality of cell movement was greatly promoted by the presence of calreticulin. Calreticulin expression was also found to have a dramatic effect on the phosphorylation state of serine 636 of IRS-1, such that phosphorylation of IRS-1 on serine 636 increased radically in the absence of calreticulin. Most importantly, treatment of cells with the RhoA/ROCK inhibitor, Y-27632, which among its many effects also inhibited serine 636 phosphorylation of IRS-1, had profound effects on cell-substratum adhesion, in that it suppressed focal contacts, induced extensive close contacts, and increased the strength of adhesion. The latter effect, while counterintuitive, can be explained by the close contacts comprising labile bonds but in large numbers. In addition, the lability of bonds in close contacts would permit fast locomotion. An interesting and novel finding is that Y-27632 treatment of MEFs releases them from contact inhibition of locomotion, as evidenced by the invasion of a cell's underside by the thin lamellae and filopodia of a cell in close apposition.


Asunto(s)
Calreticulina/biosíntesis , Movimiento Celular/genética , Proteínas Sustrato del Receptor de Insulina/genética , Transducción de Señal/genética , Amidas/administración & dosificación , Animales , Calreticulina/genética , Adhesión Celular/efectos de los fármacos , Adhesión Celular/genética , Movimiento Celular/efectos de los fármacos , Inhibición de Contacto/efectos de los fármacos , Retículo Endoplásmico/genética , Fibroblastos/efectos de los fármacos , Adhesiones Focales/efectos de los fármacos , Adhesiones Focales/genética , Humanos , Proteínas Sustrato del Receptor de Insulina/antagonistas & inhibidores , Ratones , Fosforilación , Piridinas/administración & dosificación
12.
Aging (Albany NY) ; 6(12): 1010-8, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25585637

RESUMEN

The most physiological type of cell cycle arrest - namely, contact inhibition in dense culture - is the least densely studied. Despite cell cycle arrest, confluent cells do not become senescent. We recently described that mTOR (target of rapamycin) is inactive in contact-inhibited cells. Therefore, conversion from reversible arrest to senescence (geroconversion) is suppressed. I this Perspective, we further extended the gerosuppression model. While causing senescence in regular cell density, etoposide failed to cause senescence in contact-inhibited cells. A transient reactivation of mTOR favored geroconversion in etoposide-treated confluent cells. Like p21, p16 did not cause senescence in high cell density. We discuss that suppression of geroconversion in confluent and contact-inhibited cultures mimics gerosuppression in the organism. We confirmed that levels of p-S6 were low in murine tissues in the organism compared with mouse embryonic fibroblasts in cell culture, whereas p-Akt was reciprocally high in the organism.


Asunto(s)
Puntos de Control del Ciclo Celular , Senescencia Celular , Inhibición de Contacto , Fibroblastos/fisiología , Animales , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular , Proliferación Celular , Senescencia Celular/efectos de los fármacos , Inhibición de Contacto/efectos de los fármacos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Etopósido/farmacología , Fibroblastos/efectos de los fármacos , Fibroblastos/enzimología , Humanos , Ratones , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Quinasas S6 Ribosómicas/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Factores de Tiempo
13.
J Cell Mol Med ; 17(12): 1619-31, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24118792

RESUMEN

The production of hydrogen peroxide (H2 O2 ) drives tumourigenesis in ulcerative colitis (UC). Recently, we showed that H2 O2 activates DNA damage checkpoints in human colonic epithelial cells (HCEC) through c-Jun N-terminal Kinases (JNK) that induces p21(WAF1) . Moreover, caspases circumvented the G1/S and intra-S checkpoints, and cells accumulated in G2/M. The latter observation raised the question of whether repeated H2 O2 exposures alter JNK activation, thereby promoting a direct passage of cells from G2/M arrest to driven cell cycle progression. Here, we report that increased proliferation of repeatedly H2 O2 -exposed HCEC cells (C-cell cultures) was associated with (i) increased phospho-p46 JNK, (ii) decreased total JNK and phospho-p54 JNK and (iii) p21(WAF1) down-regulation. Altered JNK activation and p21(WAF1) down-regulation were accompanied by defects in maintaining G2/M and mitotic spindle checkpoints through adaptation, as well as by apoptosis resistance following H2 O2 exposure. This may cause increased proliferation of C-cell cultures, a defining initiating feature in the inflammation-carcinoma pathway in UC. We further suggest that dysregulated JNK activation is attributed to a non-apoptotic function of caspases, causing checkpoint adaptation in C-cell cultures. Additionally, loss of cell-contact inhibition and the overcoming of senescence, hallmarks of cancer, contributed to increased proliferation. Furthermore, there was evidence that p54 JNK inactivation is responsible for loss of cell-contact inhibition. We present a cellular model of UC and suggest a sinusoidal pattern of proliferation, which is triggered by H2 O2 -induced reactive oxygen species generation, involving an interplay between JNK activation/inactivation, p21(WAF1) , c-Fos, c-Jun/phospho-c-Jun, ATF2/phospho-ATF2, ß-catenin/TCF4-signalling, c-Myc, CDK6 and Cyclin D2, leading to driven cell cycle progression.


Asunto(s)
Ciclo Celular/efectos de los fármacos , Colitis Ulcerosa/patología , Peróxido de Hidrógeno/farmacología , Modelos Biológicos , Caspasas/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Inhibición de Contacto/efectos de los fármacos , Ciclina D2/metabolismo , Quinasa 6 Dependiente de la Ciclina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/enzimología , Humanos , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Factores de Transcripción/metabolismo
14.
PLoS One ; 8(8): e71490, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23940764

RESUMEN

Pulmonary artery endothelial cells (PAEC) in an intact vessel are continually exposed to serum, but unless injured, do not proliferate, constrained by confluence. In contrast, pulmonary artery smooth muscle cells (PASMC) attain, and maintain, confluence in the presence of minimal serum, protected from serum's stimulatory effects except when the endothelial barrier becomes more permeable. We hypothesized therefore, that confluent PASMC may be less constrained by contact inhibition in the presence of serum than PAEC and tested this idea by exposing confluent non-transformed human PAEC and PASMC to media containing increasing concentrations of fetal bovine serum (FBS) and determining cell growth over 7 days. PAEC that had attained confluence in low serum did not proliferate even when exposed to 5% serum, the highest concentration tested. In contrast, PASMC that attained confluence in low serum did proliferate once serum levels were increased, an effect that was dose dependent. Consistent with this observation, PASMC had more BrdU incorporation and a greater percentage of cells in S phase in 5% compared to 0.2% FBS, whereas no such difference was seen in PAEC. These results suggest that confluent human PAEC are resistant to the stimulatory effects of serum, whereas confluent PASMC can proliferate when serum levels are increased, an effect mediated in part by differences in phosphoinositide 3-kinase activation. This observation may be relevant to understanding the PASMC hyperplasia observed in humans and animals with pulmonary hypertension in which changes in endothelial permeability due to hypoxia or injury expose the underlying smooth muscle to serum.


Asunto(s)
Inhibición de Contacto , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/citología , Arteria Pulmonar/citología , Suero/fisiología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Inhibición de Contacto/efectos de los fármacos , Medios de Cultivo/farmacología , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/fisiología , Humanos , Músculo Liso Vascular/fisiología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/fisiología , Proteína Oncogénica v-akt/metabolismo , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/fisiología , Proteína de Retinoblastoma/metabolismo
15.
Colloids Surf B Biointerfaces ; 108: 358-65, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23584362

RESUMEN

Copper nanoparticle based clay composite has been synthesized by in situ reduction of a copper ammonium complex ion and characterized by different analytical instruments. The copper nanoparticles were both intercalated and adsorbed on the surface with diameters of <5nm (for intercalated) and 25-30nm (for adsorbed). The composite showed good stability for over 3 months in air. Excellent antimicrobial activity of the composite was observed on Escherichia coli, Staphylococcus aureus, Pseudomonas aeruginosa and Enterococcus faecalis with mortality rates >90% after 12h. Cellular membrane damage permeated by direct attachment of the composite and indirect damage caused by released copper ion are the primary sources of antibacterial action. Cytotoxicity measurements showed minimal adverse effect on the two human cell lines beyond the M.B.C. value for the microorganisms studied. In the present form the clay composite shows good promise for use in therapeutic applications.


Asunto(s)
Antibacterianos/síntesis química , Bentonita/química , Cobre/química , Nanopartículas del Metal/química , Nanopartículas/química , Antibacterianos/farmacología , Cationes Bivalentes , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Inhibición de Contacto/efectos de los fármacos , Enterococcus faecalis/efectos de los fármacos , Enterococcus faecalis/crecimiento & desarrollo , Escherichia coli/efectos de los fármacos , Escherichia coli/crecimiento & desarrollo , Humanos , Nanopartículas del Metal/ultraestructura , Pruebas de Sensibilidad Microbiana , Microscopía Electrónica de Rastreo , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/crecimiento & desarrollo , Espectroscopía Infrarroja por Transformada de Fourier , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/crecimiento & desarrollo
16.
Arch Toxicol ; 87(3): 491-503, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23085979

RESUMEN

The aryl hydrocarbon receptor (AhR) contributes to the control of cell-to-cell communication, cell adhesion, migration or proliferation. In the present study, we investigated the regulation of connexin43 (Cx43) and Cx43-mediated gap junctional intercellular communication (GJIC) during the AhR-dependent disruption of contact inhibition in non-tumorigenic liver epithelial cells. The contact inhibition of cell proliferation is a process restricting the cell division of confluent non-transformed cells, which is frequently abolished in cancer cells; however, the mechanisms contributing to its disruption are still only partially understood. Disruption of contact inhibition, which was induced by toxic AhR ligands 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) or polycyclic aromatic hydrocarbons in epithelial WB-F344 cells, reduced Cx43 protein levels, possibly via enhanced proteasomal degradation, significantly decreased the amount of gap junction plaques and downregulated GJIC, in an AhR-dependent manner. Although both intracellular and membrane Cx43 pools were markedly reduced in cells released from contact inhibition by TCDD, siRNA-mediated Cx43 knock-down was not sufficient to stimulate proliferation in contact-inhibited cells. Our data suggest that downregulation of Cx43/GJIC in non-transformed epithelial cells is an inherent part of disruption of contact inhibition, which occurs at the post-transcriptional level. This process runs in parallel with alterations of other forms of cell-to-cell communication, thus suggesting that toxic AhR agonists may simultaneously abrogate contact inhibition and reduce GJIC, two essential mechanisms linked to deregulation of cell-to-cell communication during tumor promotion and progression.


Asunto(s)
Carcinógenos/toxicidad , Comunicación Celular/efectos de los fármacos , Conexina 43/metabolismo , Inhibición de Contacto/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Uniones Comunicantes/efectos de los fármacos , Hígado/efectos de los fármacos , Receptores de Hidrocarburo de Aril/agonistas , Transducción de Señal/efectos de los fármacos , Animales , Benzo(a)Antracenos/toxicidad , Línea Celular , Proliferación Celular , Transformación Celular Neoplásica/inducido químicamente , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Conexina 43/genética , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Células Epiteliales/metabolismo , Células Epiteliales/patología , Fluorenos/toxicidad , Uniones Comunicantes/metabolismo , Uniones Comunicantes/patología , Técnicas de Silenciamiento del Gen , Indoles/farmacología , Ligandos , Hígado/metabolismo , Hígado/patología , Neoplasias Hepáticas/inducido químicamente , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Floroglucinol/análogos & derivados , Floroglucinol/farmacología , Fosforilación , Dibenzodioxinas Policloradas/toxicidad , Complejo de la Endopetidasa Proteasomal/metabolismo , Interferencia de ARN , Ratas , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/metabolismo , Factores de Tiempo , Transfección
17.
Proc Natl Acad Sci U S A ; 109(52): 21480-5, 2012 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-23236156

RESUMEN

Contact-dependent growth inhibition (CDI) systems encode polymorphic toxin/immunity proteins that mediate competition between neighboring bacterial cells. We present crystal structures of CDI toxin/immunity complexes from Escherichia coli EC869 and Burkholderia pseudomallei 1026b. Despite sharing little sequence identity, the toxin domains are structurally similar and have homology to endonucleases. The EC869 toxin is a Zn(2+)-dependent DNase capable of completely degrading the genomes of target cells, whereas the Bp1026b toxin cleaves the aminoacyl acceptor stems of tRNA molecules. Each immunity protein binds and inactivates its cognate toxin in a unique manner. The EC869 toxin/immunity complex is stabilized through an unusual ß-augmentation interaction. In contrast, the Bp1026b immunity protein exploits shape and charge complementarity to occlude the toxin active site. These structures represent the initial glimpse into the CDI toxin/immunity network, illustrating how sequence-diverse toxins adopt convergent folds yet retain distinct binding interactions with cognate immunity proteins. Moreover, we present visual demonstration of CDI toxin delivery into a target cell.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/toxicidad , Toxinas Bacterianas/química , Toxinas Bacterianas/toxicidad , Inhibición de Contacto/efectos de los fármacos , Inmunidad/efectos de los fármacos , Proteínas Bacterianas/inmunología , Toxinas Bacterianas/inmunología , Burkholderia pseudomallei/genética , Burkholderia pseudomallei/inmunología , Burkholderia pseudomallei/metabolismo , Cristalografía por Rayos X , ADN/metabolismo , Endonucleasas/metabolismo , Escherichia coli/genética , Escherichia coli/inmunología , Escherichia coli/metabolismo , Modelos Moleculares , Familia de Multigenes/genética , Estructura Secundaria de Proteína
18.
PLoS One ; 7(5): e36864, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22590627

RESUMEN

Contact-inhibition ubiquitously exists in non-transformed cells and explains the poor regenerative capacity of in vivo human retinal pigment epithelial cells (RPE) during aging, injury and diseases. RPE injury or degeneration may unlock mitotic block mediated by contact inhibition but may also promote epithelial-mesenchymal transition (EMT) contributing to retinal blindness. Herein, we confirmed that EMT ensued in post-confluent ARPE-19 cells when contact inhibition was disrupted with EGTA followed by addition of EGF and FGF-2 because of activation of canonical Wnt and Smad/ZEB signaling. In contrast, knockdown of p120-catenin (p120) unlocked such mitotic block by activating p120/Kaiso, but not activating canonical Wnt and Smad/ZEB signaling, thus avoiding EMT. Nuclear BrdU labeling was correlated with nuclear release of Kaiso through p120 nuclear translocation, which was associated with activation of RhoA-ROCK signaling, destabilization of microtubules. Prolonged p120 siRNA knockdown followed by withdrawal further expanded RPE into more compact monolayers with a normal phenotype and a higher density. This new strategy based on selective activation of p120/Kaiso but not Wnt/ß-catenin signaling obviates the need of using single cells and the risk of EMT, and may be deployed to engineer surgical grafts containing RPE and other tissues.


Asunto(s)
Cateninas/metabolismo , Núcleo Celular/metabolismo , Inhibición de Contacto/fisiología , Transición Epitelial-Mesenquimal/fisiología , Transducción de Señal/fisiología , Factores de Transcripción/metabolismo , Transporte Activo de Núcleo Celular/efectos de los fármacos , Transporte Activo de Núcleo Celular/fisiología , Línea Celular , Inhibición de Contacto/efectos de los fármacos , Factor de Crecimiento Epidérmico/metabolismo , Factor de Crecimiento Epidérmico/farmacología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Factor 2 de Crecimiento de Fibroblastos/farmacología , Humanos , Transducción de Señal/efectos de los fármacos , Catenina delta
19.
J Cell Sci ; 125(Pt 15): 3636-48, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22505615

RESUMEN

Contact inhibition ubiquitously exists in non-transformed cells that are in contact with neighboring cells. This phenomenon explains the poor regenerative capacity of in vivo human corneal endothelial cells during aging, injury and surgery. This study demonstrated that the conventional approach of expanding human corneal endothelial cells by disrupting contact inhibition with EDTA followed by bFGF activated canonical Wnt signaling and lost the normal phenotype to endothelial-mesenchymal transition, especially if TGFß1 was added. By contrast, siRNA against p120 catenin (CTNND1) also uniquely promoted proliferation of the endothelial cells by activating trafficking of p120 catenin to the nucleus, thus relieving repression by nuclear Kaiso. This nuclear p120-catenin-Kaiso signaling is associated with activation of RhoA-ROCK signaling, destabilization of microtubules and inhibition of Hippo signaling, but not with activation of Wnt-ß-catenin signaling. Consequently, proliferating human corneal endothelial cells maintained a hexagonal shape, with junctional expression of N-cadherin, ZO-1 and Na(+)/K(+)-ATPase. Further expansion of human corneal endothelial monolayers with a normal phenotype and a higher density was possible by prolonging treatment with p120 catenin siRNA followed by its withdrawal. This new strategy of perturbing contact inhibition by selective activation of p120-catenin-Kaiso signaling without disrupting adherent junction could be used to engineer surgical grafts containing normal human corneal endothelial cells to meet a global corneal shortage and for endothelial keratoplasties.


Asunto(s)
Uniones Adherentes/fisiología , Cateninas/fisiología , Inhibición de Contacto/fisiología , Córnea/citología , Endotelio Corneal/citología , Mitosis/fisiología , Uniones Adherentes/efectos de los fármacos , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Cateninas/genética , Cateninas/metabolismo , Adhesión Celular/efectos de los fármacos , Adhesión Celular/fisiología , Procesos de Crecimiento Celular/efectos de los fármacos , Procesos de Crecimiento Celular/fisiología , Células Cultivadas , Inhibición de Contacto/efectos de los fármacos , Córnea/efectos de los fármacos , Ácido Edético/farmacología , Endotelio Corneal/efectos de los fármacos , Endotelio Corneal/ultraestructura , Transición Epitelial-Mesenquimal/efectos de los fármacos , Factor 2 de Crecimiento de Fibroblastos/farmacología , Humanos , Persona de Mediana Edad , Mitosis/efectos de los fármacos , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/metabolismo , Transfección , Factor de Crecimiento Transformador beta1/farmacología , Proteínas Wnt/metabolismo , Adulto Joven , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Catenina delta
20.
BMC Cancer ; 9: 464, 2009 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-20038293

RESUMEN

BACKGROUND: Studies of the role of the cytokine macrophage-migration-inhibitory-factor (MIF) in malignant tumors have revealed its stimulating influence on cell-cycle progression, angiogenesis and anti-apoptosis. RESULTS: Here we show that in vitro targeting MIF in cultures of human malignant glioblastoma cells by either antisense plasmid introduction or anti-MIF antibody treatment reduced the growth rates of tumor cells. Of note is the marked decrease of proliferation under confluent and over-confluent conditions, implying a role of MIF in overcoming contact inhibition. Several proteins involved in contact inhibition including p27, p21, p53 and CEBPalpha are upregulated in the MIF antisense clones indicating a restoration of contact inhibition in the tumor cells. Correspondingly, we observed a marked increase in MIF mRNA and protein content under higher cell densities in LN18 cells. Furthermore, we showed the relevance of the enzymatic active site of MIF for the proliferation of glioblastoma cells by using the MIF-tautomerase inhibitor ISO-1. CONCLUSION: Our study adds another puzzle stone to the role of MIF in tumor growth and progression by showing the importance of MIF for overcoming contact inhibition.


Asunto(s)
Inhibición de Contacto/genética , Glioblastoma/patología , Oxidorreductasas Intramoleculares/genética , Factores Inhibidores de la Migración de Macrófagos/genética , Antineoplásicos/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Inhibición de Contacto/efectos de los fármacos , Progresión de la Enfermedad , Técnicas de Silenciamiento del Gen , Marcación de Gen , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Oxidorreductasas Intramoleculares/antagonistas & inhibidores , Oxidorreductasas Intramoleculares/inmunología , Oxidorreductasas Intramoleculares/metabolismo , Factores Inhibidores de la Migración de Macrófagos/antagonistas & inhibidores , Factores Inhibidores de la Migración de Macrófagos/inmunología , Factores Inhibidores de la Migración de Macrófagos/metabolismo , ARN Interferente Pequeño/farmacología , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...