Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Nucleic Acids Res ; 48(5): 2621-2642, 2020 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-31863590

RESUMEN

Transposable elements (TEs) comprise a large proportion of long non-coding RNAs (lncRNAs). Here, we employed CRISPR to delete a short interspersed nuclear element (SINE) in Malat1, a cancer-associated lncRNA, to investigate its significance in cellular physiology. We show that Malat1 with a SINE deletion forms diffuse nuclear speckles and is frequently translocated to the cytoplasm. SINE-deleted cells exhibit an activated unfolded protein response and PKR and markedly increased DNA damage and apoptosis caused by dysregulation of TDP-43 localization and formation of cytotoxic inclusions. TDP-43 binds stronger to Malat1 without the SINE and is likely 'hijacked' by cytoplasmic Malat1 to the cytoplasm, resulting in the depletion of nuclear TDP-43 and redistribution of TDP-43 binding to repetitive element transcripts and mRNAs encoding mitotic and nuclear-cytoplasmic regulators. The SINE promotes Malat1 nuclear retention by facilitating Malat1 binding to HNRNPK, a protein that drives RNA nuclear retention, potentially through direct interactions of the SINE with KHDRBS1 and TRA2A, which bind to HNRNPK. Losing these RNA-protein interactions due to the SINE deletion likely creates more available TDP-43 binding sites on Malat1 and subsequent TDP-43 aggregation. These results highlight the significance of lncRNA TEs in TDP-43 proteostasis with potential implications in both cancer and neurodegenerative diseases.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteostasis/genética , ARN Largo no Codificante/genética , Elementos de Nucleótido Esparcido Corto/genética , Apoptosis , Línea Celular , Citoplasma/metabolismo , Daño del ADN , Estrés del Retículo Endoplásmico , Activación Enzimática , Dosificación de Gen , Ribonucleoproteína Heterogénea-Nuclear Grupo K/metabolismo , Humanos , Mitosis , Modelos Biológicos , Transporte de Proteínas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Eliminación de Secuencia/genética , eIF-2 Quinasa
2.
Development ; 141(7): 1562-71, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24598160

RESUMEN

Disruptions in polarity and mitotic spindle orientation contribute to the progression and evolution of tumorigenesis. However, little is known about the molecular mechanisms regulating these processes in vivo. Here, we demonstrate that Polo-like kinase 2 (Plk2) regulates mitotic spindle orientation in the mammary gland and that this might account for its suggested role as a tumor suppressor. Plk2 is highly expressed in the mammary gland and is required for proper mammary gland development. Loss of Plk2 leads to increased mammary epithelial cell proliferation and ductal hyperbranching. Additionally, a novel role for Plk2 in regulating the orientation of the mitotic spindle and maintaining proper cell polarity in the ductal epithelium was discovered. In support of a tumor suppressor function for Plk2, loss of Plk2 increased the formation of lesions in multiparous glands. Collectively, these results demonstrate a novel role for Plk2 in regulating mammary gland development.


Asunto(s)
Polaridad Celular/genética , Glándulas Mamarias Animales/crecimiento & desarrollo , Proteínas Serina-Treonina Quinasas/fisiología , Huso Acromático/genética , Animales , Células Cultivadas , Epitelio/metabolismo , Epitelio/fisiología , Femenino , Regulación del Desarrollo de la Expresión Génica , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones SCID , Proteínas Serina-Treonina Quinasas/genética
3.
PLoS Genet ; 8(7): e1002840, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22911650

RESUMEN

Pregnancy-induced noncoding RNA (PINC) and retinoblastoma-associated protein 46 (RbAp46) are upregulated in alveolar cells of the mammary gland during pregnancy and persist in alveolar cells that remain in the regressed lobules following involution. The cells that survive involution are thought to function as alveolar progenitor cells that rapidly differentiate into milk-producing cells in subsequent pregnancies, but it is unknown whether PINC and RbAp46 are involved in maintaining this progenitor population. Here, we show that, in the post-pubertal mouse mammary gland, mPINC is enriched in luminal and alveolar progenitors. mPINC levels increase throughout pregnancy and then decline in early lactation, when alveolar cells undergo terminal differentiation. Accordingly, mPINC expression is significantly decreased when HC11 mammary epithelial cells are induced to differentiate and produce milk proteins. This reduction in mPINC levels may be necessary for lactation, as overexpression of mPINC in HC11 cells blocks lactogenic differentiation, while knockdown of mPINC enhances differentiation. Finally, we demonstrate that mPINC interacts with RbAp46, as well as other members of the polycomb repressive complex 2 (PRC2), and identify potential targets of mPINC that are differentially expressed following modulation of mPINC expression levels. Taken together, our data suggest that mPINC inhibits terminal differentiation of alveolar cells during pregnancy to prevent abundant milk production and secretion until parturition. Additionally, a PRC2 complex that includes mPINC and RbAp46 may confer epigenetic modifications that maintain a population of mammary epithelial cells committed to the alveolar fate in the involuted gland.


Asunto(s)
Diferenciación Celular , Glándulas Mamarias Animales/metabolismo , Embarazo/metabolismo , ARN no Traducido/metabolismo , Proteínas Represoras/metabolismo , Proteína 7 de Unión a Retinoblastoma/metabolismo , Animales , Femenino , Técnicas de Silenciamiento del Gen , Glándulas Mamarias Animales/citología , Ratones , Ratones Endogámicos BALB C , Proteínas del Grupo Polycomb , ARN no Traducido/genética , Ratas
4.
J Clin Invest ; 133(24)2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-37874652

RESUMEN

Protein synthesis is frequently dysregulated in cancer and selective inhibition of mRNA translation represents an attractive cancer therapy. Here, we show that therapeutically targeting the RNA helicase eIF4A with zotatifin, the first-in-class eIF4A inhibitor, exerts pleiotropic effects on both tumor cells and the tumor immune microenvironment in a diverse cohort of syngeneic triple-negative breast cancer (TNBC) mouse models. Zotatifin not only suppresses tumor cell proliferation but also directly repolarizes macrophages toward an M1-like phenotype and inhibits neutrophil infiltration, which sensitizes tumors to immune checkpoint blockade. Mechanistic studies revealed that zotatifin reprograms the tumor translational landscape, inhibits the translation of Sox4 and Fgfr1, and induces an interferon (IFN) response uniformly across models. The induction of an IFN response is partially due to the inhibition of Sox4 translation by zotatifin. A similar induction of IFN-stimulated genes was observed in breast cancer patient biopsies following zotatifin treatment. Surprisingly, zotatifin significantly synergizes with carboplatin to trigger DNA damage and an even heightened IFN response, resulting in T cell-dependent tumor suppression. These studies identified a vulnerability of eIF4A in TNBC, potential pharmacodynamic biomarkers for zotatifin, and provide a rationale for new combination regimens consisting of zotatifin and chemotherapy or immunotherapy as treatments for TNBC.


Asunto(s)
Antineoplásicos , Neoplasias de la Mama Triple Negativas , Animales , Ratones , Humanos , Interferones , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología , Línea Celular Tumoral , Antineoplásicos/farmacología , Proliferación Celular , Microambiente Tumoral
5.
bioRxiv ; 2023 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-37808840

RESUMEN

Protein synthesis is frequently dysregulated in cancer and selective inhibition of mRNA translation represents an attractive cancer therapy. Here, we show that therapeutically targeting the RNA helicase eIF4A by Zotatifin, the first-in-class eIF4A inhibitor, exerts pleiotropic effects on both tumor cells and the tumor immune microenvironment in a diverse cohort of syngeneic triple-negative breast cancer (TNBC) mouse models. Zotatifin not only suppresses tumor cell proliferation but also directly repolarizes macrophages towards an M1-like phenotype and inhibits neutrophil infiltration, which sensitizes tumors to immune checkpoint blockade. Mechanistic studies revealed that Zotatifin reprograms the tumor translational landscape, inhibits the translation of Sox4 and Fgfr1, and induces an interferon response uniformly across models. The induction of an interferon response is partially due to the inhibition of Sox4 translation by Zotatifin. A similar induction of interferon-stimulated genes was observed in breast cancer patient biopsies following Zotatifin treatment. Surprisingly, Zotatifin significantly synergizes with carboplatin to trigger DNA damage and an even heightened interferon response resulting in T cell-dependent tumor suppression. These studies identified a vulnerability of eIF4A in TNBC, potential pharmacodynamic biomarkers for Zotatifin, and provide a rationale for new combination regimens comprising Zotatifin and chemotherapy or immunotherapy as treatments for TNBC.

6.
Cancer Res Commun ; 1(3): 178-193, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-35156101

RESUMEN

Polo-like kinase (PLK) family members play important roles in cell cycle regulation. The founding member PLK1 is oncogenic and preclinically validated as a cancer therapeutic target. Paradoxically, frequent loss of chromosome 5q11-35 which includes PLK2 is observed in basal-like breast cancer. In this study, we found that PLK2 was tumor suppressive in breast cancer, preferentially in basal-like and triple-negative breast cancer (TNBC) subtypes. Knockdown of PLK1 rescued phenotypes induced by PLK2-loss both in vitro and in vivo. We also demonstrated that PLK2 directly interacted with PLK1 at prometaphase through the kinase but not the polo-box domains of PLK2, suggesting PLK2 functioned at least partially through the interaction with PLK1. Furthermore, an improved treatment response was seen in both Plk2-deleted/low mouse preclinical and PDX TNBC models using the PLK1 inhibitor volasertib alone or in combination with carboplatin. Re-expression of PLK2 in an inducible PLK2-null mouse model reduced the therapeutic efficacy of volasertib. In summary, this study delineates the effects of chromosome 5q loss in TNBC that includes PLK2, the relationship between PLK2 and PLK1, and how this may render PLK2-deleted/low tumors more sensitive to PLK1 inhibition in combination with chemotherapy.


Asunto(s)
Neoplasias de la Mama Triple Negativas , Humanos , Ratones , Animales , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Genes Supresores de Tumor , Biomarcadores , Proteínas Serina-Treonina Quinasas/genética
7.
J Biol Chem ; 284(34): 22815-24, 2009 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-19542223

RESUMEN

Lactogenic hormone regulation of beta-casein gene expression in mammary epithelial cells provides an excellent model in which to study the mechanisms by which steroid and peptide hormone signaling control gene expression. Prolactin- and glucocorticoid-mediated induction of beta-casein gene expression involves two principal regulatory regions, a proximal promoter and a distal enhancer located in the mouse approximately -6 kb upstream of the transcription start site. Using a chromosome conformation capture assay and quantitative real time PCR, we demonstrate that a chromatin loop is created in conjunction with the recruitment of specific transcription factors and p300 in HC11 mammary epithelial cells. Stimulation with both prolactin and hydrocortisone is required for the induction of these long range interactions between the promoter and enhancer, and no DNA looping was observed in nontreated cells or cells treated with each of the hormones separately. The lactogenic hormone-induced interaction between the proximal promoter and distal enhancer was confirmed in hormone-treated primary three-dimensional mammary acini cultures. In addition, the developmental regulation of DNA looping between the beta-casein regulatory regions was observed in lactating but not in virgin mouse mammary glands. Furthermore, beta-casein mRNA induction and long range interactions between these regulatory regions were inhibited in a progestin-dependent manner following stimulation with prolactin and hydrocortisone in HC11 cells expressing human PR-B. Collectively, these data suggest that the communication between these regulatory regions with intervening DNA looping is a crucial step required to both create and maintain active chromatin domains and regulate transcription.


Asunto(s)
Caseínas/genética , Glucocorticoides/farmacología , Histona Desacetilasas/metabolismo , Prolactina/farmacología , Factor de Transcripción STAT5/metabolismo , Factor de Transcripción YY1/metabolismo , Animales , Técnicas de Cultivo de Célula , Línea Celular , Células Cultivadas , Cromatina/genética , Cromatina/metabolismo , Inmunoprecipitación de Cromatina , Elementos de Facilitación Genéticos/genética , Regulación del Desarrollo de la Expresión Génica/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Humanos , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/metabolismo , Ratones , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas/genética , Unión Proteica/efectos de los fármacos
8.
Cancer Res ; 78(15): 4229-4240, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29844125

RESUMEN

Cooperativity between WNT and FGF signaling is well documented in embryonic development and cancer progression, but the molecular mechanisms underlying this cross-talk remain elusive. In this study, we interrogated the dynamics of RNA levels, ribosome occupancy, and protein expression as a function of inducible FGF signaling in mouse mammary glands with constitutive WNT hyperactivation. Multiomics correlation analysis revealed a substantial discrepancy between RNA and ribosome occupancy levels versus protein levels. However, this discrepancy decreased as cells became premalignant and dynamically responded to FGF signaling, implicating the importance of stringent gene regulation in nontransformed cells. Analysis of individual genes demonstrated that acute FGF hyperactivation increased translation of many stem cell self-renewal regulators, including WNT signaling components, and decreased translation of genes regulating cellular senescence. WNT pathway components translationally upregulated by FGF signaling had long and structured 5' UTRs with a high frequency of polypurine sequences, several of which harbored (CGG)4 motifs that can fold into either stable G-quadruplexes or other stable secondary structures. The FGF-mediated increase in translation of WNT pathway components was compromised by silvestrol, an inhibitor of EIF4A that clamps EIF4A to polypurine sequences to block 43S scanning and inhibits its RNA-unwinding activity important for translation initiation. Moreover, silvestrol treatment significantly delayed FGF-WNT-driven tumorigenesis. Taken together, these results suggest that FGF signaling selectively enhances translation of structured mRNAs, particularly WNT signaling components, and highlight their vulnerability to inhibitors that target the RNA helicase EIF4A.Significance: The RNA helicase EIF4A may serve as a therapeutic target for breast cancers that require FGF and WNT signaling. Cancer Res; 78(15); 4229-40. ©2018 AACR.


Asunto(s)
Regiones no Traducidas 5'/genética , Factor 4A Eucariótico de Iniciación/genética , Biosíntesis de Proteínas/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Vía de Señalización Wnt/genética , Regiones no Traducidas 5'/efectos de los fármacos , Animales , Ratones , Biosíntesis de Proteínas/efectos de los fármacos , ARN Helicasas/genética , ARN Mensajero/genética , Ribosomas/efectos de los fármacos , Ribosomas/genética , Triterpenos/farmacología , Vía de Señalización Wnt/efectos de los fármacos
9.
Mol Endocrinol ; 20(10): 2355-68, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16772529

RESUMEN

Lactogenic hormone regulation of beta-casein gene expression in mammary epithelial cells provides an excellent system in which to perform kinetic studies of chromatin remodeling and transcriptional activation. Using HC11 cells as a model, we have investigated the effects of prolactin (Prl) and glucocorticoids both singly and in combination at different time points after hormone treatment. Using chromatin immunoprecipitation analysis, we have determined the dynamics of assembly and disassembly of signal transducer and activator of transcription 5, glucocorticoid receptor, CCAAT enhancer binding protein beta, and Ying Yang-1 at the hormonally activated beta-casein proximal promoter as well as the distal mouse beta-casein enhancer located approximately -6 kb upstream of the transcription start site. Prl alone resulted in a rapid recruitment of both signal transducer and activator of transcription 5 and histone deacetylase 1 to the beta-casein promoter and enhancer, and reciprocally the dissociation of Ying Yang-1 from the proximal promoter. In addition, we have examined the recruitment of coactivator p300 and determined chromatin acetylation status as a function of hormonal treatment. Finally, we have established the time course of RNA polymerase II and phospho-RNA polymerase II accumulation at the beta-casein promoter and enhancer after stimulation with hydrocortisone and Prl. Although glucocorticoids alone led to a rapid increase in histone H3 acetylation, treatment with both hormones was required for stable association of p300 and phospho-RNA polymerase II at both the promoter and enhancer. Collectively, these data suggest a model for the assembly of a multiprotein complex that helps to define how the signaling pathways controlled by these lactogenic hormones are integrated to regulate beta-casein gene expression.


Asunto(s)
Caseínas/metabolismo , Ensamble y Desensamble de Cromatina/fisiología , Regulación de la Expresión Génica/fisiología , Glándulas Mamarias Animales/metabolismo , Prolactina/metabolismo , Receptores de Glucocorticoides/metabolismo , Transducción de Señal/fisiología , Factores de Transcripción/metabolismo , Animales , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Caseínas/genética , Línea Celular , Inmunoprecipitación de Cromatina , Cartilla de ADN , Elementos de Facilitación Genéticos/genética , Glándulas Mamarias Animales/citología , Ratones , Modelos Biológicos , Regiones Promotoras Genéticas/genética , ARN Polimerasa II/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT5/metabolismo , Factor de Transcripción YY1/metabolismo
10.
Mol Endocrinol ; 16(12): 2675-91, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12456789

RESUMEN

Targeted deletion of the bZIP transcription factor, CCAAT/enhancer binding protein-beta (C/EBPbeta), was shown previously to result in aberrant ductal morphogenesis and decreased lobuloalveolar development, accompanied by an altered pattern of progesterone receptor (PR) expression. Here, similar changes in the level and pattern of prolactin receptor (PrlR) expression were observed while screening for differentially expressed genes in C/EBPbeta(null) mice. PR patterning was also altered in PrlR(null) mice, as well as in mammary tissue transplants from both PrlR(null) and signal transducer and activator of transcription (Stat) 5a/b-deficient mice, with concomitant defects in hormone-induced proliferation. Down-regulation of PR and activation of Stat5 phosphorylation were seen after estrogen and progesterone treatment in both C/EBPbeta(null) and wild-type mice, indicating that these signaling pathways were functional, despite the failure of steroid hormones to induce proliferation. IGF binding protein-5, IGF-II, and insulin receptor substrate-1 all displayed altered patterns and levels of expression in C/EBPbeta(null) mice, suggestive of a change in the IGF signaling axis. In addition, small proline-rich protein (SPRR2A), a marker of epidermal differentiation, and keratin 6 were misexpressed in the mammary epithelium of C/EBPbeta(null) mice. Together, these data suggest that C/EBPbeta is a master regulator of mammary epithelial cell fate and that the correct spatial pattern of PR and PrlR expression is a critical determinant of hormone-regulated cell proliferation.


Asunto(s)
Regulación de la Expresión Génica , Glándulas Mamarias Animales/crecimiento & desarrollo , Proteínas de la Leche , Receptores de Progesterona/genética , Receptores de Prolactina/genética , Animales , Proteína beta Potenciadora de Unión a CCAAT/deficiencia , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteína beta Potenciadora de Unión a CCAAT/fisiología , Diferenciación Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Proteínas Ricas en Prolina del Estrato Córneo , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Células Epidérmicas , Células Epiteliales/citología , Estradiol/administración & dosificación , Femenino , Hibridación in Situ , Proteínas Sustrato del Receptor de Insulina , Proteína 5 de Unión a Factor de Crecimiento Similar a la Insulina/genética , Factor II del Crecimiento Similar a la Insulina/genética , Glándulas Mamarias Animales/química , Glándulas Mamarias Animales/trasplante , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfoproteínas/genética , Fosforilación , Fosfotirosina/metabolismo , Progesterona/administración & dosificación , Prolactina/farmacología , Precursores de Proteínas/genética , ARN Mensajero/análisis , Receptores de Progesterona/análisis , Receptores de Progesterona/fisiología , Receptores de Prolactina/deficiencia , Receptores de Prolactina/fisiología , Factor de Transcripción STAT5 , Transducción de Señal , Transactivadores/deficiencia , Transactivadores/genética , Transactivadores/fisiología
11.
Mol Endocrinol ; 25(6): 955-68, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21527503

RESUMEN

Differentiated HC-11 cells ectopically expressing progesterone receptor (PR) were used to explore the molecular mechanisms by which progesterone suppresses ß-casein gene transcription induced by prolactin (PRL) and glucocorticoids in the mammary gland. As detected by chromatin immunoprecipitation assays, treatment of cells with the progestin agonist R5020 induced a rapid recruitment (5 min) of PR to the proximal promoter (-235 bp) and distal enhancer (-6 kb upstream of transcription start site) of ß-casein. PR remained bound for 4 h and was dissociated by 24 h after treatment. Despite efficient binding, the hormone agonist-occupied PR did not stimulate transcription of the ß-casein gene. Recruitment of signal transducer and activator of transcription 5a, glucocorticoid receptor, and the CCAAT enhancer binding protein ß to the enhancer and proximal promoter of ß-casein induced by PRL and glucocorticoids was blocked by progestin cotreatment, whereas PR binding was induced under these conditions. PRL/glucocorticoid-induced histone acetylation and the recruitment of the coactivator p300 and RNA polymerase II required for gene activation were also inhibited by progestin. In addition, progestin prevented dissociation of the corepressors Yin and Yang 1 and histone deacetylase 3 from the promoter, and demethylation of lysine 9 of histone 3 induced by PRL and glucocorticoids. These studies are consistent with the conclusion that progesterone interferes with PRL/glucocorticoid induction of ß-casein transcription by a physical interaction of PR with the promoter and enhancer that blocks assembly of a transcriptional activation complex and dissociation of corepressors and promotes repressive chromatin modifications. These studies define a novel mechanism of steroid receptor-mediated transcriptional repression of a physiologically important gene in mammary gland development and differentiation.


Asunto(s)
Caseínas/genética , Cromatina/metabolismo , Elementos de Facilitación Genéticos , Células Epiteliales/metabolismo , Glándulas Mamarias Animales/citología , Regiones Promotoras Genéticas , Receptores de Progesterona/metabolismo , Transcripción Genética , Animales , Caseínas/metabolismo , Línea Celular , Cromatina/genética , Células Epiteliales/efectos de los fármacos , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Hidrocortisona/farmacología , Glándulas Mamarias Animales/efectos de los fármacos , Glándulas Mamarias Animales/metabolismo , Ratones , Progestinas/farmacología , Prolactina/farmacología , Promegestona/farmacología , Multimerización de Proteína , Receptores de Glucocorticoides/antagonistas & inhibidores , Receptores de Glucocorticoides/metabolismo , Receptores de Progesterona/agonistas , Factor de Transcripción STAT5/genética , Factor de Transcripción STAT5/metabolismo , Transcripción Genética/efectos de los fármacos , Activación Transcripcional/efectos de los fármacos
12.
J Biol Chem ; 278(19): 17218-27, 2003 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-12621061

RESUMEN

Stat5 is activated by a broad spectrum of cytokines, as well as non-receptor tyrosine kinases, such as Src. In this study, the DNA binding properties of the two closely related Stat5 proteins, Stat5a and Stat5b, induced either by prolactin (Prl) or by Src were analyzed by electrophoretic mobility shift assays using several different Stat5 binding sites. Src-induced Stat5b-DNA binding complexes consistently displayed a slightly faster mobility than those induced by Prl, as well as differences in their ability to be supershifted by anti-Stat5 antibodies. IP-Westerns performed using specific antibodies directed at the N and C termini of Stat5b suggested that depending on the activating stimulus, Stat5b exhibited different conformations, which influenced antibody accessibility at its C terminus. These conformational differences may in part be due to differential effects of Prl and Src on Stat5b tyrosine phosphorylation, since Src induced several additional sites of tyrosine phosphorylation of Stat5b at residues other than Tyr-699, including Tyr-724 and Tyr-679. The latter Tyr-679 is conserved in all mammalian Stat5bs, but is not present in Stat5a. A Stat 5bY679F mutant induced by Src kinase exhibited an altered pattern of nuclear localization as compared with wild-type Stat5b. Furthermore, this mutation inhibited v-Src-induced cyclin D1-luciferase reporter activity in transient transfection assays performed in Stat5a/b-deficient MEFs, suggesting that Tyr-679 phosphorylation may play a role in v-Src induced proliferation. Thus, depending on the signal transduction pathway responsible for activation, different conformations of activated Stat5 may result in selective biological responses.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas de la Leche , Prolactina/metabolismo , Transducción de Señal , Transactivadores/metabolismo , Familia-src Quinasas/metabolismo , Proteínas de Unión al ADN/química , Activación Enzimática , Células HeLa , Humanos , Conformación Proteica , Factor de Transcripción STAT5 , Relación Estructura-Actividad , Transactivadores/química , Proteínas Supresoras de Tumor
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA