Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Oncotarget ; 8(33): 55361-55373, 2017 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-28903425

RESUMEN

Pasireotide is a somatostatin analog (SSA) that targets somatostatin receptor subtype 1 (SST1), SST2, SST3, and SST5 with a high affinity. Pasireotide has a better antisecretory effect in acromegaly, Cushing's disease, and neuroendocrine tumors than octreotide. In this study, we compared the effects of pasireotide to those of octreotide in vitro on meningioma primary cell cultures, both alone and in combination with the mTOR inhibitor everolimus. Significant mRNA expression levels of SST1, SST2, and SST5 were observed in 40.5%, 100%, and 35% of meningioma samples, respectively. Pasireotide had a significantly stronger inhibitory effect on cell proliferation than octreotide. The effect of pasireotide, but not of octreotide, was significantly stronger in the group expressing the highest level of SST1 mRNA. Combined treatment with pasireotide and everolimus induced a higher reduction in cell viability than that with octreotide plus everolimus. Moreover, pasireotide decreased Akt phosphorylation and reversed everolimus-induced Akt hyperphosphorylation to a higher degree than octreotide. Using 4E-BP1 siRNA (si4E-BP), we demonstrated that 4E-BP1 protein silencing significantly reversed the response to everolimus, both alone and in combination with SSAs. Moreover, si4E-BP completely reversed the inhibition of cyclin D1 expression level and the increase in p27kip1 induced by SSAs, both alone and in combination with everolimus. Our results strongly support the need for further studies on the combination of pasireotide and everolimus in medical therapy for meningiomas.

2.
J Neurosurg ; 127(3): 660-669, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27982767

RESUMEN

OBJECTIVE Meningiomas express somatostatin receptor subtype 2 (SST2), which is targeted by the somatostatin analog octreotide. However, to date, using somatostatin analog therapy for the treatment of these tumors in clinical practice has been debated. This study aims to clarify the in vitro effects of octreotide on meningiomas for precise clinical applications. METHODS The effects of octreotide were analyzed in a large series of 80 meningiomas, including 31 World Health Organization (WHO) Grade II and 4 WHO Grade III tumors, using fresh primary cell cultures to study the impact on cell viability, apoptosis, and signal transduction pathways. RESULTS SST2 mRNA was detected in 100% of the tested meningiomas at levels similar to those observed in other SST2-expressing tumors, neuroendocrine tumors, or pituitary adenomas. Octreotide significantly decreased cell proliferation in 88% of meningiomas but did not induce cell death. On average, cell proliferation was more inhibited in the meningioma group expressing a high level of SST2 than in the low-SST2 group. Moreover, octreotide response was positively correlated to the level of merlin protein and inversely correlated to the level of phosphorylated p70-S6 kinase, a downstream effector of the PI3K/Akt/mammalian target of rapamycin (mTOR) pathway. Octreotide inhibited Akt phosphorylation and activated tyrosine phosphatase without impacting the extracellular regulated kinase (ERK) pathway. CONCLUSIONS Octreotide acts exclusively as an antiproliferative agent and does not promote apoptosis in meningioma in vitro. Therefore, in vivo, octreotide is likely to limit tumor growth rather than induce tumor shrinkage. A meta-analysis of the literature reveals an interest in octreotide for the treatment of WHO Grade I tumors, particularly those in the skull base for which the 6-month progression-free survival level reached 92%. Moreover, somatostatin analogs, which are well-tolerated drugs, could be of interest for use as co-targeting therapies for aggressive meningiomas.


Asunto(s)
Antineoplásicos Hormonales/uso terapéutico , Meningioma/tratamiento farmacológico , Octreótido/uso terapéutico , Antineoplásicos Hormonales/farmacología , Proliferación Celular/efectos de los fármacos , Correlación de Datos , Humanos , Técnicas In Vitro , Octreótido/farmacología , Células Tumorales Cultivadas
3.
Endocr Relat Cancer ; 23(7): 509-19, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27267119

RESUMEN

Pegvisomant (PEG), an antagonist of growth hormone (GH)-receptor (GHR), normalizes insulin-like growth factor 1 (IGF1) oversecretion in most acromegalic patients unresponsive to somatostatin analogs (SSAs) and/or uncontrolled by transsphenoidal surgery. The residual GH-secreting tumor is therefore exposed to the action of circulating PEG. However, the biological effect of PEG at the pituitary level remains unknown. To assess the impact of PEG in vitro on the hormonal secretion (GH and prolactin (PRL)), proliferation and cellular viability of eight human GH-secreting tumors in primary cultures and of the rat somatolactotroph cell line GH4C1. We found that the mRNA expression levels of GHR were characterized in 31 human GH-secreting adenomas (0.086 copy/copy ß-Gus) and the GHR was identified by immunocytochemistry staining. In 5/8 adenomas, a dose-dependent inhibition of GH secretion was observed under PEG with a maximum of 38.2±17% at 1µg/mL (P<0.0001 vs control). A dose-dependent inhibition of PRL secretion occurred in three mixed GH/PRL adenomas under PEG with a maximum of 52.8±11.5% at 10µg/mL (P<0.0001 vs control). No impact on proliferation of either human primary tumors or GH4C1 cell line was observed. We conclude that PEG inhibits the secretion of GH and PRL in primary cultures of human GH(/PRL)-secreting pituitary adenomas without effect on cell viability or cell proliferation.


Asunto(s)
Adenoma/metabolismo , Adenoma Hipofisario Secretor de Hormona del Crecimiento/metabolismo , Hormona del Crecimiento/metabolismo , Hormona de Crecimiento Humana/análogos & derivados , Prolactina/metabolismo , Adulto , Anciano , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Femenino , Hormona de Crecimiento Humana/farmacología , Humanos , Janus Quinasa 2/metabolismo , Masculino , Persona de Mediana Edad , Ratas , Receptores de Somatotropina/genética , Receptores de Somatotropina/metabolismo , Factor de Transcripción STAT5/metabolismo , Células Tumorales Cultivadas , Adulto Joven
4.
J Neurooncol ; 124(1): 33-43, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26015296

RESUMEN

Treatment for recurrent and aggressive meningiomas remains an unmet medical need in neuro-oncology, and chemotherapy exhibits limited clinical activity, if any. Merlin expression, encoded by the NF2 gene, is lost in a majority of meningiomas, and merlin is a negative regulator of mTORC1. The sst2 somatostatin receptor, targeted by octreotide, is highly expressed in meningiomas. To investigate new therapeutic strategies, we evaluated the activity of everolimus (mTOR inhibitor), BKM-120 and BEZ-235 (new Pi3K/Akt/mTOR inhibitors), octreotide and a combined treatment (octreotide plus everolimus), on cell proliferation, signaling pathways, and cell cycle proteins, respectively. The in vitro study was conducted on human meningioma primary cells extracted from fresh tumors, allowing the assessment of somatostatin analogs at the concentration levels used in patients. The results were correlated to WHO grades. Further, everolimus decreased cell viability of human meningiomas, but concomitantly, induced Akt activation, reducing the antiproliferative effect of the drug. The new Pi3K inhibitors were not more active than everolimus alone, limiting their clinical relevance. In contrast, a clear cooperative inhibitory effect of octreotide and everolimus was observed on cell proliferation in all tested meningiomas, including WHO grades II-III. Octreotide not only reversed everolimus-induced Akt phosphorylation but also displayed additive and complementary effects with everolimus on downstream proteins involved in translation (4EB-P1), and controlling cell cycle (p27Kip1 and cyclin D1). We have demonstrated a co-operative action between everolimus and octreotide on cell proliferation in human meningiomas, including aggressive ones, establishing the basis for a clinical trial.


Asunto(s)
Antineoplásicos/administración & dosificación , Everolimus/administración & dosificación , Neoplasias Meníngeas/tratamiento farmacológico , Meningioma/tratamiento farmacológico , Octreótido/administración & dosificación , Adulto , Anciano , Anciano de 80 o más Años , Aminopiridinas/administración & dosificación , Ciclo Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Quimioterapia Combinada , Femenino , Humanos , Imidazoles/administración & dosificación , Masculino , Persona de Mediana Edad , Morfolinas/administración & dosificación , Neurofibromina 2/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-akt/metabolismo , Quinolinas/administración & dosificación , Receptores de Somatostatina/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/antagonistas & inhibidores
5.
J Clin Endocrinol Metab ; 99(12): E2463-71, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25272306

RESUMEN

CONTEXT: The ghrelin receptor GHS-R1a is highly expressed in human somatotroph adenomas and exhibits unusually high basal signaling activity. In humans, the suppression of this constitutive activity by mutation induces a short stature. OBJECTIVE: Using a GHS-R1a inverse agonist, modified substance P (MSP), we explored the role of GHS-R1a constitutive activity in GH hypersecretion from somatotroph adenomas and as a putative therapeutic target. DESIGN: The effects of MSP were assessed on GH secretion from 19 human somatotroph tumors in vitro. Moreover, these effects were compared with those of octreotide (somatostatin receptor subtype 2 [sst2] agonist) and with the combination of both drugs. Expression and localization of GHS-R1a and sst2 were studied. RESULTS: For all tumors, MSP inhibited GH secretion in a dose-dependent manner from 13 to 64%. Moreover, MSP enhanced octreotide-induced GH inhibition. For five tumors, the effects of combined MSP plus octreotide treatment were significantly higher than the sum of effects of each drug alone. MSP increased the membrane localization of GHS-R1a and of microdomains colocalizing sst2-GHS-R1a, highlighting the cooperation between the two drugs. CONCLUSIONS: The GHS-R1a inverse agonist could open new therapeutic options for acromegalic patients, particularly patients partially sensitive to octreotide whose GH secretion is not completely controlled by the sst2 agonist.


Asunto(s)
Adenoma/tratamiento farmacológico , Antineoplásicos Hormonales/uso terapéutico , Adenoma Hipofisario Secretor de Hormona del Crecimiento/tratamiento farmacológico , Receptores de Ghrelina/agonistas , Receptores de Ghrelina/metabolismo , Somatostatina/análogos & derivados , Somatostatina/uso terapéutico , Adulto , Anciano , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Femenino , Hormona de Crecimiento Humana/antagonistas & inhibidores , Hormona de Crecimiento Humana/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Octreótido/farmacología , Receptores de Somatostatina/metabolismo , Sustancia P/uso terapéutico , Adulto Joven
6.
Am J Primatol ; 76(1): 56-64, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24038166

RESUMEN

Fagot and Paleressompoulle [Fagot and Paleressompoulle (2009) Behav Res Methods 41: 396-404] described a new automated learning device for monkeys (ALDM) to test the cognitive functions of nonhuman primates within their social groups. However, the impact of the ALDM procedure on animal well-being needs to be investigated. The present study assessed the consequences of ALDM testing on the behavioral repertoire of Guinea baboons (Papio papio) and their stress levels as inferred from measurements of saliva cortisol. Accessibility to ALDM test computers reduced the number of resting periods as well as the number of stereotypies. Lower cortisol levels were also found during ALDM testing. These findings and others demonstrate that ALDM testing has a positive impact on animal well-being and can be considered as a means for behavioral enrichment in captive primates.


Asunto(s)
Animales de Laboratorio/fisiología , Conducta Animal/fisiología , Cognición , Hidrocortisona/metabolismo , Aprendizaje , Papio papio/fisiología , Bienestar del Animal , Animales , Masculino , Saliva/química , Estrés Fisiológico , Factores de Tiempo
7.
Endocr Relat Cancer ; 20(5): 753-66, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23940012

RESUMEN

Germline aryl hydrocarbon receptor interacting protein (AIP) gene mutations confer a predisposition to pituitary adenoma (PA), predominantly GH-secreting (GH-PA). As recent data suggest a role for AIP in the pathogenesis of sporadic GH-PA and their response to somatostatin analogues (SSA), the expression of AIP and its partner, aryl hydrocarbon receptor (AHR), was determined by semiquantitative immunohistochemistry scoring in 62 sporadic GH-PA (37 treated with SSA preoperatively). The influence of Gsp status was studied in a subset of tumours (n=39, 14 Gsp(+)) and six GH-PA were available for primary cultures. AIP and AHR were detected in most cases, with a positive correlation between AIP and cytoplasmic AHR (P=0.012). Low AIP expression was significantly more frequent in untreated vs SSA-treated tumours (44.0 vs 20.5%, P=0.016). AHR expression or localisation did not differ between the two groups. Similarly, in vitro octreotide induced a median twofold increase in AIP expression (range 1.2-13.9, P=0.027) in GH-PA. In SSA-treated tumours, the AIP score was significantly higher in the presence of preoperative IGF1 decrease or tumour shrinkage (P=0.008 and P=0.014 respectively). In untreated tumours, low AIP expression was significantly associated with invasiveness (P=0.028) and suprasellar extension (P=0.019). The only effect of Gsp status was a significantly lower nuclear AHR score in Gsp(+) vs Gsp(-) tumours (P=0.025), irrespective of SSA. In conclusion, AIP is involved in the aggressiveness of sporadic GH-PA, regardless of Gsp status, and AIP up-regulation in SSA-treated tumours is associated with a better preoperative response, with no clear role for AHR.


Asunto(s)
Antineoplásicos Hormonales/farmacología , Adenoma Hipofisario Secretor de Hormona del Crecimiento/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Octreótido/farmacología , Somatostatina/farmacología , Adolescente , Adulto , Anciano , Femenino , Subunidades alfa de la Proteína de Unión al GTP/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Persona de Mediana Edad , Mutación , Receptores de Hidrocarburo de Aril/metabolismo , Somatostatina/análogos & derivados , Adulto Joven
8.
Mol Cell Endocrinol ; 355(1): 106-13, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22348806

RESUMEN

OBJECTIVE: As prolactinomas fail to respond to dopamine agonist (DA) in 10-20% of cases, we hypothesized that somatostatin subtype 2 receptor (sst2) overexpression in DA-resistant prolactinomas may enhance suppression of prolactine (PRL) using chimeric agonist (dopastatin) that simultaneously binds sst2 and the dopamine subtype 2 receptor (D2DR). DESIGN AND METHODS: PRL suppression by octreotide, sst5 agonist, sst2-D2DR agonist (BIM-23A760 dopastatin) and cabergoline was assessed in primary cultures of seven DA-resistant prolactinomas overexpressing sst2. RESULTS: sst2 was effectively overexpressed via adenoviral expression in prolactinomas (38.1±7.4 vs. 0.1±0.1 copy/copy ß-Gus) and induced octreotide sst2-mediated PRL suppression that remained lower than that induced by DA. BIM-23A760 inhibited PRL similarly to cabergoline both in the control and sst2-expressing cells. Antagonist experiments confirmed predominant dopaminergic effect in dopastatin activity. CONCLUSION: sst2 was successfully overexpressed in prolactinomas. However BIM-23A760 was unable to enhance PRL suppression underlining a predominant dopaminergic contribution in its action.


Asunto(s)
Agonistas de Dopamina/farmacología , Neoplasias Hipofisarias/tratamiento farmacológico , Prolactina/antagonistas & inhibidores , Prolactinoma/tratamiento farmacológico , Receptores de Somatostatina/metabolismo , Adenoviridae , Adolescente , Adulto , Cabergolina , Dopamina/análogos & derivados , Dopamina/metabolismo , Dopamina/farmacología , Ergolinas/farmacología , Femenino , Vectores Genéticos , Humanos , Masculino , Octreótido/farmacología , Neoplasias Hipofisarias/metabolismo , Neoplasias Hipofisarias/patología , Cultivo Primario de Células , Prolactina/genética , Prolactinoma/metabolismo , Prolactinoma/patología , Receptores de Dopamina D2/metabolismo , Receptores de Somatostatina/genética , Somatostatina/análogos & derivados , Somatostatina/farmacología , Transfección
9.
Endocrinology ; 152(10): 3884-92, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21810944

RESUMEN

Nonfunctioning pituitary adenomas (NFPA; gonadotroph derived), even not inducing hormonal hypersecretion, cause significant morbidity by compression neighboring structures. No effective and specific medical methods are available so far for treating these tumors. The pituitary homeobox 2 (PITX2) gene is a member of the bicoid-like homeobox transcription factor family, which is involved in the Wnt/Dvl/ß-catenin pathway. PITX2 is overexpressed in NFPA. PITX2 mutations are known to be responsible for Axenfield Rieger syndrome, a genetic disorder in which pituitary abnormalities have been detected. The R91P mutant identified in Axenfeld Rieger syndrome is a dominant-negative factor, which is able to block the expression of several pituitary genes activated by PITX2. To better understand the role of Pitx2 on gonadotroph tumorigenesis and to explore new approach for inhibiting tumoral growth, the R91P mutant was transferred via a lentiviral vector in tumoral gonadotroph cells of two kinds: the αT3-1 cell line and human adenoma cells. R91P mutant and small interfering RNA directed against Pitx2 both decreased the viability of αT3-1 cells via an apoptotic mechanism involving the activation of executioner caspase. Similar effects of the R91P mutant were observed on human gonadotroph cells in primary culture. Therefore, Pitx2 overexpression may play an antiapoptotic role during NFPA tumorigenesis.


Asunto(s)
Adenoma/etiología , Gonadotrofos/patología , Proteínas de Homeodominio/fisiología , Neoplasias Hipofisarias/etiología , Factores de Transcripción/fisiología , Adenoma/patología , Animales , Apoptosis , Supervivencia Celular , Proteínas de Homeodominio/antagonistas & inhibidores , Proteínas de Homeodominio/genética , Humanos , Lentivirus/genética , Ratones , Neoplasias Hipofisarias/patología , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética , Proteína del Homeodomínio PITX2
10.
Biol Cell ; 103(11): 519-29, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21787362

RESUMEN

BACKGROUND INFORMATION: Previous studies have reported that cross-talk between integrins may be an important regulator of integrin-ligand binding and subsequent signalling events that control a variety of cell functions in many tissues. We previously demonstrated that αvß5/ß6 integrin represses α2ß1-dependent cell migration. The αv subunits undergo an endoproteolytic cleavage by protein convertases, whose role in tumoral invasion has remained controversial. RESULTS: Inhibition of convertases by the convertase inhibitor α1-PDX (α1-antitrypsin Portland variant), leading to the cell-surface expression of an uncleaved form of the αv integrin, stimulated cell migration toward type I collagen. Under convertase inhibition, α2ß1 engagement led to enhanced phosphorylation of both FAK (focal adhesion kinase) and MAPK (mitogen-activated protein kinase). This outside-in signalling stimulation was associated with increased levels of activated ß1 integrin located in larger than usual focal-adhesion structures and a cell migration that was independent of the PI3K (phosphoinositide 3-kinase)/Akt (also called protein kinase B) pathway. CONCLUSIONS: The increase in cell migration observed upon convertases inhibition appears to be due to the up-regulation of ß1 integrins and to their location in larger focal-adhesion structures. The endoproteolytic cleavage of αv subunits is necessary for αvß5/ß6 integrin to control α2ß1 function and could thus play an essential role in colon cancer cell migration.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Integrina alfa2beta1/metabolismo , Integrina alfaV/metabolismo , Integrinas/metabolismo , Receptores de Vitronectina/metabolismo , Adhesión Celular , Movimiento Celular , Colágeno Tipo I/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Humanos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , alfa 1-Antitripsina/biosíntesis
11.
PLoS One ; 5(4): e10124, 2010 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-20405031

RESUMEN

Integrins are essential protagonists of the complex multi-step process of angiogenesis that has now become a major target for the development of anticancer therapies. We recently reported and characterized that MVL-PLA2, a novel phospholipase A2 from Macrovipera lebetina venom, exhibited anti-integrin activity. In this study, we show that MVL-PLA2 also displays potent anti-angiogenic properties. This phospholipase A2 inhibited adhesion and migration of human microvascular-endothelial cells (HMEC-1) in a dose-dependent manner without being cytotoxic. Using Matrigel and chick chorioallantoic membrane assays, we demonstrated that MVL-PLA2, as well as its catalytically inactivated form, significantly inhibited angiogenesis both in vitro and in vivo. We have also found that the actin cytoskeleton and the distribution of alphav beta3 integrin, a critical regulator of angiogenesis and a major component of focal adhesions, were disturbed after MVL-PLA2 treatment. In order to further investigate the mechanism of action of this protein on endothelial cells, we analyzed the dynamic instability behavior of microtubules in living endothelial cells. Interestingly, we showed that MVL-PLA2 significantly increased microtubule dynamicity in HMEC-1 cells by 40%. We propose that the enhancement of microtubule dynamics may explain the alterations in the formation of focal adhesions, leading to inhibition of cell adhesion and migration.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Adhesiones Focales/metabolismo , Microtúbulos/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Fosfolipasas A2/farmacología , Inhibidores de la Angiogénesis , Animales , Adhesión Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Células Endoteliales/citología , Endotelio Vascular/citología , Humanos , Venenos de Serpiente/farmacología
12.
Matrix Biol ; 28(4): 188-93, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19351557

RESUMEN

Here, we report the purification and characterization of an acidic Asp49 phospholipase A2, named MVL-PLA2, with a molecular mass of 13,626.64 Da. The complete MVL-PLA2 cDNA was cloned from Macrovipera lebetina transmediterranea venom gland cDNA library. MVL-PLA2 possesses 122 amino acid residues, including 14 cysteines, and belongs to group II snake venom phospholipase A2 enzymes. MVL-PLA2 was not cytotoxic up to 2 muM and completely abolished cell adhesion and migration of various human tumor cells. Chemical modification with p-bromophenacyl bromide abolished the enzymatic activity of MVL-PLA2 without affecting its anti-tumor effect, suggesting the presence of 'pharmacological sites' distinct from the catalytic site in snake venom phospholipase A2. We demonstrated for the first time that the anti-tumor effect of MVL-PLA2 was mediated by alpha5beta1 and alphav-containing integrins. This finding may serve as starting point for structure-function relationship studies leading to design a new generation of specific anti-cancer drugs.


Asunto(s)
Antineoplásicos/farmacología , Fosfolipasas A2/farmacología , Venenos de Víboras/enzimología , Viperidae/metabolismo , Secuencia de Aminoácidos , Animales , Antígenos de Neoplasias , Antineoplásicos/química , Antineoplásicos/aislamiento & purificación , Dominio Catalítico/efectos de los fármacos , Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Fibrosarcoma/patología , Humanos , Integrina alfa5beta1/antagonistas & inhibidores , Integrina alfaVbeta3/antagonistas & inhibidores , Integrinas/antagonistas & inhibidores , Melanoma/patología , Datos de Secuencia Molecular , Proteínas de Neoplasias/antagonistas & inhibidores , Fosfolipasas A2/química , Fosfolipasas A2/aislamiento & purificación , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Relación Estructura-Actividad , Venenos de Víboras/química , Venenos de Víboras/aislamiento & purificación , Venenos de Víboras/farmacología
13.
Exp Cell Res ; 315(11): 1840-9, 2009 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-19328197

RESUMEN

Crosstalk between integrins is involved in the regulation of various cell functions including cell migration. Here we identify the interplay between the integrins alphavbeta5/beta6 and alpha2beta1 during cell migration toward type I collagen. Human colon cancer cell lines HT29-D4 and SW480 were used as cell models. To improve our understanding of the consequences of alphavbeta5/beta6 function on alpha2beta1, we decreased the expression of alphav integrins by either siRNA or lysosomal targeting strategies or inhibited their function using, as antagonists, blocking antibodies or disintegrins. In all cases, we observed a greatly enhanced alpha2beta1 integrin-dependent cell migration associated with focal adhesion rearrangements and increased outside-in signaling as demonstrated by elevated phosphorylation of focal adhesion kinase and MAPKinase (ERK1 and ERK2). The alphavbeta5/beta6-dependent limitation of alpha2beta1 function could be overridden by TS2/16, an activating anti-beta1 antibody. Interestingly, compared to control cells, the pharmacological inhibition of PI3Kinase or the siRNA-mediated knockdown of AKT had little effect on the high alpha2beta1-mediated cell migration observed in the absence of alphav integrins or following activation of alpha2beta1 integrins by the TS2/16. These results suggest that integrins alphavbeta5/beta6 repress alpha2beta1 possibly by interfering with their activation process and thereby modify the cell signaling regulation of alpha2beta1-mediated migration.


Asunto(s)
Movimiento Celular/fisiología , Integrina alfa2beta1/fisiología , Integrinas/antagonistas & inhibidores , Proteína Oncogénica v-akt/antagonistas & inhibidores , Inhibidores de las Quinasa Fosfoinosítidos-3 , Receptores de Vitronectina/antagonistas & inhibidores , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/fisiología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Colágeno Tipo I/metabolismo , Adhesiones Focales , Humanos , Integrina alfa2beta1/antagonistas & inhibidores , Integrina alfa2beta1/genética , Integrinas/genética , Integrinas/fisiología , Proteína Oncogénica v-akt/genética , Proteína Oncogénica v-akt/fisiología , Fosfatidilinositol 3-Quinasas/fisiología , Inhibidores de Proteínas Quinasas/farmacología , ARN Interferente Pequeño/genética , Receptores de Vitronectina/genética , Receptores de Vitronectina/fisiología , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA