Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell Mol Life Sci ; 76(5): 961-975, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30506415

RESUMEN

The tubulin cytoskeleton is one of the main components of the cytoarchitecture and is involved in several cellular functions. Here, we examine the interplay between Listeria monocytogenes (Lm) and the tubulin cytoskeleton upon cellular infection. We show that non-polymeric tubulin is present throughout Lm actin comet tails and, to a less extent, in actin clouds. Moreover, we demonstrate that stathmin, a regulator of microtubule dynamics, is also found in these Lm-associated actin structures and is required for tubulin recruitment. Depletion of host stathmin results in longer comets containing less F-actin, which may be correlated with higher levels of inactive cofilin in the comet, thus suggesting a defect on local F-actin dynamics. In addition, intracellular bacterial speed is significantly reduced in stathmin-depleted cells, revealing the importance of stathmin/tubulin in intracellular Lm motility. In agreement, the area of infection foci and the total bacterial loads are also significantly reduced in stathmin-depleted cells. Collectively, our results demonstrate that stathmin promotes efficient cellular infection, possibly through tubulin recruitment and control of actin dynamics at Lm-polymerized actin structures.


Asunto(s)
Actinas/metabolismo , Listeria monocytogenes/patogenicidad , Estatmina/fisiología , Tubulina (Proteína)/metabolismo , Actinas/química , Animales , Línea Celular , Humanos , Ratones , Microtúbulos/fisiología , Ratas , Tubulina (Proteína)/química
2.
FASEB J ; 30(9): 3202-15, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27284003

RESUMEN

Stathmin is a prominent destabilizer of microtubules (MTs). Extensive in vitro studies have strongly suggested that stathmin could act by sequestering tubulin and/or by binding to MT tips. In cells, the molecular mechanisms of stathmin binding to tubulin and/or MTs and its implications for the MT dynamics remain unexplored. By using immunofluorescence resonance energy transfer and fluorescence recovery after photobleaching, we analyzed the ability of stathmin and its phosphorylated forms (on Ser16, -25, -38, and -63) to interact with tubulin and MTs in A549 cells. Consistent with in vitro studies, we detected stathmin-tubulin interactions at the MT plus ends and in the cytosol. Of interest, we also observed a novel pool of stathmin bound along the MT. Expression of truncated stathmin and use of MT-stabilizing taxol further showed that the C-terminal domain of stathmin is the main contributor to this binding and that the phosphorylation state of stathmin plays a role in its binding along the MT wall. Our findings demonstrate that stathmin binds directly along the MT wall. This pool of stathmin would be readily available to participate in protofilament dissociation when the moving plus end of a depolymerizing MT reaches stathmin molecules.-Nouar, R., Breuzard, G., Bastonero, S., Gorokhova, S., Barbier, P., Devred, F., Kovacic, H., Peyrot, V. Direct evidence for the interaction of stathmin along the length and the plus end of microtubules in cells.


Asunto(s)
Microtúbulos/fisiología , Estatmina/fisiología , Anticuerpos , Línea Celular Tumoral , ADN Complementario/genética , ADN Complementario/metabolismo , Regulación de la Expresión Génica/fisiología , Humanos , Immunoblotting , Paclitaxel/farmacología , Fosforilación , Moduladores de Tubulina/farmacología
3.
Hepatology ; 58(5): 1558-68, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23703790

RESUMEN

UNLABELLED: Host factors play an important role in all facets of the hepatitis C virus (HCV) life cycle and one such host factor is signal transducer and activator of transcription 3 (STAT3). The HCV core protein has been shown to directly interact with and activate STAT3, while oxidative stress generated during HCV replication in a replicon-based model also induced STAT3 activation. However, despite these findings the precise role of STAT3 in the HCV life cycle remains unknown. We have established that STAT3 is actively phosphorylated in the presence of replicating HCV. Furthermore, expression of a constitutively active form of STAT3 leads to marked increases in HCV replication, whereas, conversely, chemical inhibition and small interfering RNA (siRNA) knockdown of STAT3 leads to significant decreases in HCV RNA levels. This strongly implicates STAT3 as a proviral host factor. As STAT3 is a transcription factor, up-regulation of a distinct set of STAT3-dependent genes may create an environment that is favorable for HCV replication. However, STAT3 has recently been demonstrated to positively regulate microtubule (MT) dynamics, by way of a direct sequestration of the MT depolymerizing protein Stathmin 1 (STMN1), and we provide evidence that STAT3 may exert its effect on the HCV life cycle by way of positive regulation of MT dynamics. CONCLUSION: We have demonstrated that STAT3 plays a role in the life cycle of HCV and have clarified the role of STAT3 as a proviral host factor.


Asunto(s)
Hepacivirus/fisiología , Factor de Transcripción STAT3/fisiología , Carcinoma Hepatocelular/etiología , Línea Celular Tumoral , Humanos , Neoplasias Hepáticas/etiología , Microtúbulos/fisiología , ARN Interferente Pequeño/genética , Factor de Transcripción STAT3/antagonistas & inhibidores , Estatmina/fisiología , Replicación Viral
4.
J Immunol ; 188(11): 5421-7, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22529300

RESUMEN

Polarization of T cells involves reorientation of the microtubule organizing center (MTOC). Because activated ERK is localized at the immunological synapse, we investigated its role by showing that ERK activation is important for MTOC polarization. Suspecting that ERK phosphorylates a regulator of microtubules, we next focused on stathmin, a known ERK substrate. Our work indicates that during T cell activation, ERK is recruited to the synapse, allowing it to phosphorylate stathmin molecules near the immunological synapse. Supporting an important role of stathmin phosphorylation in T cell activation, we showed that T cell activation results in increased microtubule growth rate dependent on the presence of stathmin. The significance of this finding was demonstrated by results showing that CTLs from stathmin(-/-) mice displayed defective MTOC polarization and defective target cell cytolysis. These data implicate stathmin as a regulator of the microtubule network during T cell activation.


Asunto(s)
Polaridad Celular/inmunología , Activación de Linfocitos/inmunología , Centro Organizador de los Microtúbulos/metabolismo , Estatmina/fisiología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Animales , Aumento de la Célula , Células Cultivadas , Humanos , Células Jurkat , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Centro Organizador de los Microtúbulos/inmunología , Fosforilación/inmunología , Estatmina/deficiencia , Estatmina/metabolismo , Subgrupos de Linfocitos T/citología
5.
Acta Biochim Biophys Sin (Shanghai) ; 46(12): 1034-40, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25348735

RESUMEN

Glioma is one of the most highly angiogenic tumors, and glioma stem cells (GSCs) are responsible for resistance to chemotherapy and radiotherapy, as well as recurrence after operation. Stathmin is substantial for mitosis and plays an important role in proliferation and migration of glioma-derived endothelial cells. However, the relationship between stathmin and GSCs is incompletely understood. Here we isolated GSCs from glioma cell lines U87MG and U251, and then used siRNA targeting stathmin for silencing. We showed that silencing of stathmin suppressed the proliferation, increased the apoptosis rate, and arrested the cell cycle at G2/M phase in GSCs. Silencing of stathmin in GSCs also resulted in inhibited the migration/invasion as well as the capability of vasculogenic mimicry. The susceptibility of GSCs to temozolomide was also enhanced by stathmin silencing. Our findings suggest stathmin as a potential target in GSCs for glioma treatment.


Asunto(s)
Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Glioma/tratamiento farmacológico , Células Madre Neoplásicas/efectos de los fármacos , ARN Interferente Pequeño/genética , Estatmina/fisiología , Secuencia de Bases , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Cartilla de ADN , Silenciador del Gen , Glioma/patología , Humanos , Interferencia de ARN , Reacción en Cadena en Tiempo Real de la Polimerasa , Estatmina/genética
6.
FASEB J ; 26(9): 3862-74, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22700873

RESUMEN

Microtubule (MT) dynamics in vascular endothelium are modulated by vasoactive mediators and are critically involved in the control of endothelial cell (EC) permeability via Rho GTPase-dependent crosstalk with the actin cytoskeleton. However, the role of regulators in MT stability in these mechanisms remains unclear. This study investigated the involvement of the MT-associated protein stathmin in the mediation of agonist-induced permeability in EC cultures and vascular leak in vivo. Thrombin treatment of human pulmonary ECs induced rapid dephosphorylation and activation of stathmin. Inhibition of stathmin activity by small interfering RNA-based knockdown or cAMP-mediated phosphorylation abrogated thrombin-induced F-actin remodeling and Rho-dependent EC hyperpermeability, while expression of a phosphorylation-deficient stathmin mutant exacerbated thrombin-induced EC barrier disruption. Stathmin suppression preserved the MT network against thrombin-induced MT disassembly and release of Rho-specific guanine nucleotide exchange factor, GEF-H1. The protective effects of stathmin knockdown were observed in vivo in the mouse 2-hit model of ventilator-induced lung injury and were linked to MT stabilization and down-regulation of Rho signaling in the lung. These results demonstrate the mechanism of stathmin-dependent control of MT dynamics, Rho signaling, and permeability and suggest novel potential pharmacological interventions in the prevention of increased vascular leak via modulation of stathmin activity.


Asunto(s)
Permeabilidad de la Membrana Celular , Pulmón/citología , Microtúbulos/fisiología , Estatmina/fisiología , Animales , Endotelio/citología , Endotelio/efectos de los fármacos , Técnica del Anticuerpo Fluorescente , Técnicas de Silenciamiento del Gen , Pulmón/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Fosforilación , ARN Interferente Pequeño/genética , Respiración Artificial/efectos adversos , Estatmina/genética , Estatmina/metabolismo , Trombina/farmacología
7.
Dev Cell ; 10(4): 497-508, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16580994

RESUMEN

Cell migration within a natural context is tightly controlled, often by specific transcription factors. However, the switch from stationary to migratory behavior is poorly understood. Border cells perform a spatially and temporally controlled invasive migration during Drosophila oogenesis. Slbo, a C/EBP family transcriptional activator, is required for them to become migratory. We purified wild-type and slbo mutant border cells as well as nonmigratory follicle cells and performed comparative whole-genome expression profiling, followed by functional tests of the contributions of identified targets to migration. About 300 genes were significantly upregulated in border cells, many dependent on Slbo. Among these, the microtubule regulator Stathmin was strongly upregulated and was required for normal migration. Actin cytoskeleton regulators were also induced, including, surprisingly, a large cluster of "muscle-specific" genes. We conclude that Slbo induces multiple cytoskeletal effectors, and that each contributes to the behavioral changes in border cells.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/fisiología , Movimiento Celular/fisiología , Proteínas de Drosophila/fisiología , Perfilación de la Expresión Génica , Oogénesis/fisiología , Ovario/fisiología , Factores de Transcripción/fisiología , Transcripción Genética , Animales , Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas del Citoesqueleto/fisiología , Drosophila , Proteínas de Drosophila/genética , Femenino , Oogénesis/genética , Ovario/citología , Ovario/metabolismo , Estatmina/fisiología , Factores de Transcripción/genética , Regulación hacia Arriba
8.
Mutat Res ; 722(2): 154-64, 2011 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-20816848

RESUMEN

Stathmin/oncoprotein 18, a protein that regulates microtubule dynamics, is highly expressed in a number of tumors including leukemia, lymphoma, neuroblastoma, breast, ovarian, and prostate cancers. High stathmin levels have been associated with the development of resistance to the widely used anti-cancer drug taxol ((®)Taxol, paclitaxel). The mechanisms of stathmin-mediated taxol resistance are not well-understood at the molecular level. To better understand the role of stathmin in taxol resistance, we stably overexpressed stathmin twofold in BT549 human breast cancer cells and characterized several cell processes involved in the mechanism of action of taxol. After stable overexpression of stathmin, neither the cell doubling time nor the mitotic index was altered and the microtubule polymer mass was reduced only modestly (by 18%). Unexpectedly, microtubule dynamicity was reduced by 29% after stathmin overexpression, resulting primarily from reduction in the catastrophe frequency. Sensitivity to taxol was reduced significantly (by 44%) in a clonogenic assay, and stathmin appeared to protect the cells from the spindle-damaging effects of taxol. The results suggest that in the stably stathmin-overexpressing clones, compensatory gene expression occurred that resulted in normal rates of cell proliferation and prevented the increase in catastrophe frequency expected in response to stathmin. Stathmin overexpression protected the cells from taxol-induced abnormal mitoses, and thus induced taxol resistance. Using offgel IEF/PAGE difference gel electrophoresis, we identified a number of proteins whose expression is reduced in the taxol-resistant stathmin-overexpressing cell lines, including proteins involved in the cytoskeleton and cell structure, the stress response, protein folding, glycolysis, and catalysis.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/genética , Resistencia a Antineoplásicos/genética , Electroforesis en Gel Bidimensional/métodos , Paclitaxel/farmacología , Estatmina/fisiología , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Proliferación Celular , Femenino , Humanos , Microtúbulos/efectos de los fármacos , Mitosis , Índice Mitótico , Proteínas de Neoplasias/efectos de los fármacos , Estatmina/genética , Estatmina/metabolismo , Regulación hacia Arriba
9.
J Neurochem ; 114(5): 1498-510, 2010 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-20569302

RESUMEN

Rat pheochromocytoma PC12 cells have been widely used to investigate the neurotrophic activities of pituitary adenylate cyclase-activating polypeptide (PACAP). In particular, PACAP has been shown to promote differentiation and to inhibit apoptosis of PC12 cells. In order to identify the mechanisms mediating these effects, we sought for proteins that are phosphorylated upon PACAP treatment. High-performance liquid chromatography and 2D gel electrophoresis analysis, coupled with mass spectrometry, revealed that stathmin 1 is strongly phosphorylated within only 5 min of exposure to PACAP. Western blot experiments confirmed that PACAP induced a robust phosphorylation of stathmin 1 in a time-dependent manner. On the other hand, PACAP decreased stathmin 1 gene expression. Investigations of the signaling mechanisms known to be activated by PACAP revealed that phosphorylation of stathmin 1 was mainly mediated through the protein kinase A and mitogen-activated protein kinase pathways. Blockage of stathmin 1 expression with small interfering RNA did not affect PC12 cell differentiation induced by PACAP but reduced the ability of the peptide to inhibit caspase 3 activity and significantly decreased its neuroprotective action. Taken together, these data demonstrate that stathmin 1 is involved in the neurotrophic effect of PACAP in PC12 cells.


Asunto(s)
Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/fisiología , Estatmina/fisiología , Secuencia de Aminoácidos , Animales , Caspasa 3/metabolismo , Supervivencia Celular/fisiología , Medio de Cultivo Libre de Suero , Regulación de la Expresión Génica/fisiología , Datos de Secuencia Molecular , Células PC12 , Ratas , Estatmina/antagonistas & inhibidores , Estatmina/biosíntesis , Estatmina/genética , Factores de Tiempo
10.
Cell Mol Life Sci ; 66(20): 3263-76, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19585080

RESUMEN

The microtubule-system organizes the cytoplasm during interphase and segregates condensed chromosomes during mitosis. Four unrelated conserved proteins, XMAP215/Dis1/TOGp, MCAK, MAP4 and Op18/stathmin, have all been implicated as predominant regulators of tubulin monomer-polymer partitioning in animal cells. However, while studies employing the Xenopus egg extract model system indicate that the partitioning is largely governed by the counteractive activities of XMAP215 and MCAK, studies of human cell lines indicate that MAP4 and Op18 are the predominant regulators of the interphase microtubule-array. Here, we review functional interplay of these proteins during interphase and mitosis in various cell model systems. We also review the evidence that MAP4 and Op18 have interphase-specific, counteractive and phosphorylation-inactivated activities that govern tubulin subunit partitioning in many mammalian cell types. Finally, we discuss evidence indicating that partitioning regulation by MAP4 and Op18 may be of significance to establish cell polarity.


Asunto(s)
Polaridad Celular , Mamíferos/metabolismo , Tubulina (Proteína)/metabolismo , Animales , Línea Celular , Humanos , Interfase/fisiología , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Asociadas a Microtúbulos/fisiología , Microtúbulos/metabolismo , Modelos Biológicos , Procesamiento Proteico-Postraduccional , Estatmina/metabolismo , Estatmina/fisiología , Tubulina (Proteína)/fisiología
11.
Med Oncol ; 37(12): 114, 2020 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-33196934

RESUMEN

CA-125, coded by MUC16 gene, is responsible to many kinds of tumor metastasis. However, the related mechanism remains unclear. We have established a novel manner to reveal gallbladder cancer metastasis related to serum CA-125 levels through the C-terminal polypeptide of MUC16 gene production. MUC16 C-terminal polypeptide significantly promoted gallbladder cancer cell migration and invasion in vitro. Mass spectrum indicated that interaction of MUC16 C-terminal fragment with the C-terminal domain of stathmin1 inhibited the phosphorylation of stathmin1 to promote the combination with tubulin. Stathmin1 knockdown inhibited the migration and invasion of gallbladder cancer cells in vitro and lung metastasis in vivo induced by MUC16 C-terminal polypeptide. The high expression level of MUC16c consistent with stathmin1 was also confirmed in gallbladder cancer tissues. Our study revealed the underlying mechanism of gallbladder cancer metastasis related to CA-125 levels, which was beneficial for CA-125 in gallbladder cancer diagnose and therapy.


Asunto(s)
Antígeno Ca-125/fisiología , Neoplasias de la Vesícula Biliar/patología , Proteínas de la Membrana/fisiología , Fragmentos de Péptidos/fisiología , Estatmina/fisiología , Línea Celular Tumoral , Movimiento Celular , Transición Epitelial-Mesenquimal , Humanos , Invasividad Neoplásica , Tubulina (Proteína)/fisiología
12.
Artículo en Inglés | MEDLINE | ID: mdl-32117057

RESUMEN

Inhibition of glucagon hypersecretion from pancreatic α-cells is an appealing strategy for the treatment of diabetes. Our hypothesis is that proteins that associate with glucagon within alpha cell secretory granules will regulate glucagon secretion, and may provide druggable targets for controlling abnormal glucagon secretion in diabetes. Recently, we identified a dynamic glucagon interactome within the secretory granules of the α cell line, αTC1-6, and showed that select proteins within the interactome could modulate glucagon secretion. In the present study, we show that one of these interactome proteins, the neuronal protein stathmin-2, is expressed in αTC1-6 cells and in mouse pancreatic alpha cells, and is a novel regulator of glucagon secretion. The secretion of both glucagon and Stmn2 was significantly enhanced in response to 55 mM K+, and immunofluorescence confocal microscopy showed co-localization of stathmin-2 with glucagon and the secretory granule markers chromogranin A and VAMP-2 in αTC1-6 cells. In mouse pancreatic islets, Stathmin-2 co-localized with glucagon, but not with insulin, and co-localized with secretory pathway markers. To show a function for stathmin-2 in regulating glucagon secretion, we showed that siRNA-mediated depletion of stathmin-2 in αTC1-6 cells caused glucagon secretion to become constitutive without any effect on proglucagon mRNA levels, while overexpression of stathmin-2 completely abolished both basal and K+-stimulated glucagon secretion. Overexpression of stathmin-2 increased the localization of glucagon into the endosomal-lysosomal compartment, while depletion of stathmin-2 reduced the endosomal localization of glucagon. Therefore, we describe stathmin-2 as having a novel role as an alpha cell secretory granule protein that modulates glucagon secretion via trafficking through the endosomal-lysosomal system. These findings describe a potential new pathway for the regulation of glucagon secretion, and may have implications for controlling glucagon hypersecretion in diabetes.


Asunto(s)
Células Secretoras de Glucagón/metabolismo , Glucagón/metabolismo , Estatmina/fisiología , Animales , Células Cultivadas , Células Secretoras de Glucagón/efectos de los fármacos , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , ARN Interferente Pequeño/farmacología , Vías Secretoras/efectos de los fármacos , Vías Secretoras/genética , Estatmina/antagonistas & inhibidores
13.
Int J Cancer ; 124(5): 1020-7, 2009 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-19048596

RESUMEN

Oncoprotein 18/stathmin (Op18/stathmin) plays a crucial role in maintaining cell biological characteristics by regulating microtubule dynamics, especially entry into mitosis; phosphorylated Op18/stathmin promotes microtubule polymerization to form the mitotic spindle, which is essential for chromosome segregation and cell division. Cdc2 is a critical kinase in starting M phase events in cell-cycle progression and is a positive regulator of the cell cycle. Latent membrane protein 1 (LMP1) is an Epstein-Barr virus (EBV)-encoded oncogenic protein that is able to induce carcinogenesis via various signaling pathways. This study focused on regulation by LMP1 of Op18/stathmin signaling in nasopharyngeal carcinoma (NPC) cells and showed that LMP1 regulates Op18/stathmin signaling through cdc2 mediation, LMP1 upregulates cdc2 kinase activity, and Op18/stathmin phosphorylation promotes the interaction of cdc2 with Op18/stathmin and microtubule polymerization during mitosis, and inhibition of LMP1 expression attenuates the interaction of cdc2 and Op18/stathmin and promotes microtubule depolymerization. These results reveal a new pathway via which LMP1 regulates Op18/stathmin signaling by cdc2 mediation; this new signaling pathway not only perfects the LMP1 regulation network but also elucidates the molecular mechanism of LMP1 that leads to carcinogenesis.


Asunto(s)
Ciclina B/fisiología , Neoplasias Nasofaríngeas/patología , Transducción de Señal/fisiología , Estatmina/fisiología , Proteínas de la Matriz Viral/fisiología , Proteína Quinasa CDC2 , Ciclo Celular , Línea Celular Tumoral , Ciclina B/antagonistas & inhibidores , Quinasas Ciclina-Dependientes , ADN Catalítico/farmacología , Herpesvirus Humano 4 , Humanos , Microtúbulos/fisiología , Fosforilación
14.
Yakugaku Zasshi ; 128(4): 565-74, 2008 Apr.
Artículo en Japonés | MEDLINE | ID: mdl-18379173

RESUMEN

Successful implantation and placentation require that trophoblasts adhere to the uterine epithelium and penetrate the decidualized endometrium. However, the biochemical mechanisms of the establishment of pregnancy including these phenomena have not yet to be definitively elucidated. We have found that stathmin, a cytosolic phosphoprotein that regulates microtubule dynamics, and insulin-like growth factor-binding protein (IGFBP)-related protein 1 (IGFBP-rP1, now called IGF-binding protein 7) were highly expressed in the endometrium around the time of implantation and decidualization. In this article, we review our recent findings of the research regarding the functions of these implantation-associated proteins in endocrine physiology and pharmacology. Analysis of the expression of both factors in rodent and human uterus has revealed that both factors are crucial for the process of endometrial stromal cell differentiation.


Asunto(s)
Implantación del Embrión/genética , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/fisiología , Estatmina/fisiología , Animales , Diferenciación Celular/genética , Endometrio/citología , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Humanos , Ratones , Embarazo , Ratas , Células del Estroma/citología
15.
PLoS One ; 12(6): e0179852, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28658321

RESUMEN

PROBLEM: To reveal the effect of p53-tristetraprolin-stathmin-1 signaling on trophoblasts and recurrent spontaneous abortion (RSA). METHOD OF STUDY: Stathmin-1 (STMN1), p53, and tristetraprolin (TTP) expression in paraffin-embedded villus tissue was determined using immunohistochemistry. HTR-8/SVneo cells were treated with doxorubicin to activate p53; STMN1 and TTP levels were detected by quantitative reverse transcription-PCR and western blotting. Western blotting and immunofluorescence were used to investigate STMN1 expression after TTP overexpression or knockdown in HTR-8 cells. RESULTS: STMN1 was downregulated and p53 was upregulated in the villus tissue from patients with RSA. Doxorubicin decreased STMN1 expression but enhanced TTP expression in HTR-8 cells. In vitro, TTP overexpression inhibited STMN1 production; TTP knockdown promoted it. TTP downregulated STMN1 expression in trophoblasts by directly binding its 3' untranslated region. CONCLUSIONS: TTP modulates trophoblast function and interacts with STMN1 and p53, and is related to pregnancy outcomes.


Asunto(s)
Aborto Espontáneo/metabolismo , Transducción de Señal/fisiología , Estatmina/fisiología , Tristetraprolina/fisiología , Proteína p53 Supresora de Tumor/fisiología , Adulto , Western Blotting , Línea Celular , Doxorrubicina/farmacología , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Recurrencia , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Trofoblastos , Adulto Joven
16.
Mol Cancer Ther ; 4(12): 1821-9, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16373697

RESUMEN

Stathmin is the founding member of a family of microtubule-destabilizing proteins that regulate the dynamics of microtubule polymerization and depolymerization. Stathmin is expressed at high levels in a variety of human cancers and provides an attractive molecule to target in cancer therapies that disrupt the mitotic apparatus. We developed replication-deficient bicistronic adenoviral vectors that coexpress green fluorescent protein and ribozymes that target stathmin mRNA. The therapeutic potential of these recombinant adenoviruses was tested in an experimental androgen-independent LNCaP prostate cancer model. Adenovirus-mediated transfer of anti-stathmin ribozymes resulted in efficient transduction and marked inhibition of stathmin expression in these cells. Cells that were transduced with the anti-stathmin adenoviruses showed a dramatic dose-dependent growth inhibition. This was associated with accumulation of LNCaP cells in the G2-M phases of the cell cycle. A similar dose-dependent inhibition of clonogenic potential was also observed in cells infected with anti-stathmin adenoviruses. Morphologic and biochemical analysis of infected cells showed a marked increase in apoptosis characterized by detachment of the cells, increased chromatin condensation, activation of caspase-3, and fragmentation of internucleosomal DNA. If these findings are confirmed in vivo, it may provide an effective approach for the treatment of prostate cancer.


Asunto(s)
Neoplasias de la Próstata/patología , Estatmina/fisiología , Adenoviridae/genética , Apoptosis , Northern Blotting , Western Blotting , Línea Celular Tumoral , Vectores Genéticos , Humanos , Masculino , ARN Catalítico/inmunología , ARN Mensajero/genética , Recombinación Genética , Estatmina/genética , Estatmina/inmunología
18.
Anat Sci Int ; 90(3): 137-43, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25595671

RESUMEN

Recently several potential susceptibility genes for major psychiatric disorders (schizophrenia and major depression) such as disrupted-in-schizophrenia 1(DISC1), dysbindin and pituitary adenylate cyclase-activating polypeptide (PACAP) have been reported. DISC1 is involved in neural development directly via adhesion molecules or via its binding partners of DISC1 such as elongation protein ζ-1 (FEZ1), DISC1-binding zinc-finger protein (DBZ) and kendrin. PACAP also regulates neural development via stathmin 1 or via regulation of the DISC1-DBZ binding. Dysbindin is also involved in neural development by regulating centrosomal microtubule network formation. All such molecules examined to date are involved in neural development. Thus, these findings provide new molecular insights into the mechanisms of neural development and neuropsychiatric disorders. On the other hand, in addition to neurons, both DISC and DBZ have been detected in oligodendrocytes and implicated in regulating oligodendrocyte differentiation. DISC1 inhibits the differentiation of oligodendrocyte precursor cells into oligodendrocytes, while DBZ has a positive regulatory role in oligodendrocyte differentiation. Evidence suggesting that disturbance of oligodendrocyte development causes major depression is also described.


Asunto(s)
Depresión/genética , Proteínas Asociadas a la Distrofina/genética , Predisposición Genética a la Enfermedad/genética , Proteínas del Tejido Nervioso/genética , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/genética , Esquizofrenia/genética , Proteínas Adaptadoras Transductoras de Señales/fisiología , Proteínas de Unión a Calmodulina/fisiología , Diferenciación Celular/genética , Proteínas de Unión al ADN/fisiología , Disbindina , Proteínas Asociadas a la Distrofina/fisiología , Humanos , Proteínas del Tejido Nervioso/fisiología , Neurogénesis/genética , Oligodendroglía/citología , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/fisiología , Unión Proteica , Estatmina/fisiología , Factores de Transcripción/fisiología
19.
Leuk Res ; 38(2): 251-7, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24355524

RESUMEN

Stathmin 1 is an important cytoplasmic microtubule-destabilizing protein that plays critical roles in proliferation and accurate chromosome segregation through regulation of microtubule dynamics. High levels of Stathmin 1 expression have been reported in leukemia and solid tumors. However, Stathmin 1 has not been studied in myelodysplastic syndrome cells. We, herein, report that significantly higher Stathmin 1 levels were observed in proliferating hematopoietic cells, in high-risk MDS and acute leukemia cells. In addition, Stathmin 1 silencing in U937 and Namalwa leukemia cells reduced cell proliferation and clonogenicity. Our data suggest that Stathmin 1 expression may be related to the highly proliferative phenotype of hematopoietic cells and add new insights into the participation of Stathmin 1 in hematological malignancies.


Asunto(s)
Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/patología , Estatmina/fisiología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Proliferación Celular , Femenino , Humanos , Masculino , Persona de Mediana Edad , Invasividad Neoplásica , Fenotipo , Riesgo , Estatmina/genética , Células Tumorales Cultivadas , Células U937 , Adulto Joven
20.
Nat Commun ; 5: 4389, 2014 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-25007915

RESUMEN

Changes in the stability of microtubules regulate many biological processes, but their role in memory remains unclear. Here we show that learning causes biphasic changes in the microtubule-associated network in the hippocampus. In the early phase, stathmin is dephosphorylated, enhancing its microtubule-destabilizing activity by promoting stathmin-tubulin binding, whereas in the late phase these processes are reversed leading to an increase in microtubule/KIF5-mediated localization of the GluA2 subunit of AMPA receptors at synaptic sites. A microtubule stabilizer paclitaxel decreases or increases memory when applied at the early or late phases, respectively. Stathmin mutations disrupt changes in microtubule stability, GluA2 localization, synaptic plasticity and memory. Aged wild-type mice show impairments in stathmin levels, changes in microtubule stability and GluA2 localization. Blocking GluA2 endocytosis rescues memory deficits in stathmin mutant and aged wild-type mice. These findings demonstrate a role for microtubules in memory in young adult and aged individuals.


Asunto(s)
Envejecimiento/fisiología , Aprendizaje/fisiología , Trastornos de la Memoria/fisiopatología , Memoria/fisiología , Microtúbulos/fisiología , Estatmina/fisiología , Animales , Hipocampo/fisiología , Hipocampo/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Ratones Transgénicos , Proteínas de Microtúbulos/fisiología , Mutación/genética , Plasticidad Neuronal/fisiología , Receptores AMPA/fisiología , Transducción de Señal/fisiología , Estatmina/deficiencia , Estatmina/genética , Tubulina (Proteína)/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA