Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Nature ; 541(7638): 541-545, 2017 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-28068668

RESUMEN

Cell fate perturbations underlie many human diseases, including breast cancer. Unfortunately, the mechanisms by which breast cell fate are regulated are largely unknown. The mammary gland epithelium consists of differentiated luminal epithelial and basal myoepithelial cells, as well as undifferentiated stem cells and more restricted progenitors. Breast cancer originates from this epithelium, but the molecular mechanisms that underlie breast epithelial hierarchy remain ill-defined. Here, we use a high-content confocal image-based short hairpin RNA screen to identify tumour suppressors that regulate breast cell fate in primary human breast epithelial cells. We show that ablation of the large tumour suppressor kinases (LATS) 1 and 2 (refs 5, 6), which are part of the Hippo pathway, promotes the luminal phenotype and increases the number of bipotent and luminal progenitors, the proposed cells-of-origin of most human breast cancers. Mechanistically, we have identified a direct interaction between Hippo and oestrogen receptor-α (ERα) signalling. In the presence of LATS, ERα was targeted for ubiquitination and Ddb1-cullin4-associated-factor 1 (DCAF1)-dependent proteasomal degradation. Absence of LATS stabilized ERα and the Hippo effectors YAP and TAZ (hereafter YAP/TAZ), which together control breast cell fate through intrinsic and paracrine mechanisms. Our findings reveal a non-canonical (that is, YAP/TAZ-independent) effect of LATS in the regulation of human breast cell fate.


Asunto(s)
Mama/citología , Mama/enzimología , Diferenciación Celular , Linaje de la Célula , Receptor alfa de Estrógeno/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteínas Adaptadoras Transductoras de Señales/agonistas , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Mama/patología , Proteínas Portadoras/metabolismo , Células Cultivadas , Receptor alfa de Estrógeno/agonistas , Femenino , Genes Supresores de Tumor , Humanos , Fosfoproteínas/agonistas , Fosfoproteínas/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Serina-Treonina Quinasas/deficiencia , Proteolisis , Transducción de Señal , Factores de Transcripción , Proteínas Supresoras de Tumor/deficiencia , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas , Proteínas Señalizadoras YAP
3.
Nature ; 525(7567): 114-8, 2015 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-26266975

RESUMEN

The adult mouse mammary epithelium contains self-sustained cell lineages that form the inner luminal and outer basal cell layers, with stem and progenitor cells contributing to its proliferative and regenerative potential. A key issue in breast cancer biology is the effect of genomic lesions in specific mammary cell lineages on tumour heterogeneity and progression. The impact of transforming events on fate conversion in cancer cells of origin and thus their contribution to tumour heterogeneity remains largely elusive. Using in situ genetic lineage tracing and limiting dilution transplantation, we have unravelled the potential of PIK3CA(H1047R), one of the most frequent mutations occurring in human breast cancer, to induce multipotency during tumorigenesis in the mammary gland. Here we show that expression of PIK3CA(H1047R) in lineage-committed basal Lgr5-positive and luminal keratin-8-positive cells of the adult mouse mammary gland evokes cell dedifferentiation into a multipotent stem-like state, suggesting this to be a mechanism involved in the formation of heterogeneous, multi-lineage mammary tumours. Moreover, we show that the tumour cell of origin influences the frequency of malignant mammary tumours. Our results define a key effect of PIK3CA(H1047R) on mammary cell fate in the pre-neoplastic mammary gland and show that the cell of origin of PIK3CA(H1047R) tumours dictates their malignancy, thus revealing a mechanism underlying tumour heterogeneity and aggressiveness.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Linaje de la Célula/genética , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/patología , Células Madre Multipotentes/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Animales , Desdiferenciación Celular/genética , Transformación Celular Neoplásica/genética , Fosfatidilinositol 3-Quinasa Clase I , Femenino , Humanos , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/patología , Ratones , Células Madre Multipotentes/patología , Mutación/genética , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Fosfatidilinositol 3-Quinasas/metabolismo
4.
Proc Natl Acad Sci U S A ; 110(11): E1026-34, 2013 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-23431153

RESUMEN

The calcium-activated chloride channel anoctamin 1 (ANO1) is located within the 11q13 amplicon, one of the most frequently amplified chromosomal regions in human cancer, but its functional role in tumorigenesis has remained unclear. The 11q13 region is amplified in ∼15% of breast cancers. Whether ANO1 is amplified in breast tumors, the extent to which gene amplification contributes to ANO1 overexpression, and whether overexpression of ANO1 is important for tumor maintenance have remained unknown. We have found that ANO1 is amplified and highly expressed in breast cancer cell lines and primary tumors. Amplification of ANO1 correlated with disease grade and poor prognosis. Knockdown of ANO1 in ANO1-amplified breast cancer cell lines and other cancers bearing 11q13 amplification inhibited proliferation, induced apoptosis, and reduced tumor growth in established cancer xenografts. Moreover, ANO1 chloride channel activity was important for cell viability. Mechanistically, ANO1 knockdown or pharmacological inhibition of its chloride-channel activity reduced EGF receptor (EGFR) and calmodulin-dependent protein kinase II (CAMKII) signaling, which subsequently attenuated AKT, v-src sarcoma viral oncogene homolog (SRC), and extracellular signal-regulated kinase (ERK) activation in vitro and in vivo. Our results highlight the involvement of the ANO1 chloride channel in tumor progression and provide insights into oncogenic signaling in human cancers with 11q13 amplification, thereby establishing ANO1 as a promising target for therapy in these highly prevalent tumor types.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Canales de Cloruro/metabolismo , Cromosomas Humanos Par 11/metabolismo , Amplificación de Genes , Proteínas de Neoplasias/metabolismo , Animales , Anoctamina-1 , Apoptosis/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Línea Celular Tumoral , Supervivencia Celular/genética , Canales de Cloruro/genética , Cromosomas Humanos Par 11/genética , Activación Enzimática/genética , Receptores ErbB/genética , Receptores ErbB/metabolismo , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteínas de Neoplasias/genética , Trasplante de Neoplasias , Transducción de Señal/genética , Trasplante Heterólogo
5.
Breast Cancer Res ; 16(3): R60, 2014 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-24916766

RESUMEN

INTRODUCTION: Stromal-epithelial interactions play a fundamental role in tissue homeostasis, controlling cell proliferation and differentiation. Not surprisingly, aberrant stromal-epithelial interactions contribute to malignancies. Studies of the cellular and molecular mechanisms underlying these interactions require ex vivo experimental model systems that recapitulate the complexity of human tissue without compromising the differentiation and proliferation potentials of human primary cells. METHODS: We isolated and characterized human breast epithelial and mesenchymal precursors from reduction mammoplasty tissue and tagged them with lentiviral vectors. We assembled heterotypic co-cultures and compared mesenchymal and epithelial cells to cells in corresponding monocultures by analyzing growth, differentiation potentials, and gene expression profiles. RESULTS: We show that heterotypic culture of non-immortalized human primary breast epithelial and mesenchymal precursors maintains their proliferation and differentiation potentials and constrains their growth. We further describe the gene expression profiles of stromal and epithelial cells in co-cultures and monocultures and show increased expression of the tumor growth factor beta (TGFß) family member inhibin beta A (INHBA) in mesenchymal cells grown as co-cultures compared with monocultures. Notably, overexpression of INHBA in mesenchymal cells increases colony formation potential of epithelial cells, suggesting that it contributes to the dynamic reciprocity between breast mesenchymal and epithelial cells. CONCLUSIONS: The described heterotypic co-culture system will prove useful for further characterization of the molecular mechanisms mediating interactions between human normal or neoplastic breast epithelial cells and the stroma, and will provide a framework to test the relevance of the ever-increasing number of oncogenomic alterations identified in human breast cancer.


Asunto(s)
Mama/citología , Células Epiteliales/citología , Subunidades beta de Inhibinas/metabolismo , Células Madre Mesenquimatosas/citología , Factor de Crecimiento Transformador beta/metabolismo , Adipocitos/citología , Tejido Adiposo/citología , Técnicas de Cultivo de Célula , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Colágeno Tipo XI/biosíntesis , Femenino , Perfilación de la Expresión Génica , Humanos , Subunidades beta de Inhibinas/biosíntesis , Cadenas alfa de Integrinas/biosíntesis , Metaloproteinasa 13 de la Matriz/biosíntesis , Proteoglicanos/biosíntesis , Transducción de Señal , Proteoglicanos Pequeños Ricos en Leucina , Factor de Crecimiento Transformador beta/biosíntesis
6.
Drug Resist Updat ; 16(3-5): 68-72, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24169539

RESUMEN

The widespread hyperactivation of the PI3K/mTOR pathway in human cancer has made it a prime target for the treatment of this disease. However, a variety of resistance mechanisms involving (re)activation of the targeted pathway or of parallel survival signaling cascades have limited the clinical success of inhibitors targeting PI3K and/or mTOR. Recent studies delineated new crosstalks between PI3K, HER2, JAK2 and IL-8 signaling, which can explain the limited efficacy of PI3K blockade when inhibitors of this pathway are used as single agents. In this review, we summarize molecular mechanisms of resistance to inhibitors of the PI3K/mTOR pathway, provide an outline of new connections between crucial oncogenic signaling pathways, and discuss the potential of new combination therapy approaches to overcome resistance.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Resistencia a Antineoplásicos , Janus Quinasa 2/antagonistas & inhibidores , Modelos Biológicos , Inhibidores de las Quinasa Fosfoinosítidos-3 , Receptor ErbB-2/antagonistas & inhibidores , Animales , Mama/efectos de los fármacos , Mama/inmunología , Mama/metabolismo , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/metabolismo , Sinergismo Farmacológico , Femenino , Humanos , Inmunomodulación/efectos de los fármacos , Interleucina-8/antagonistas & inhibidores , Interleucina-8/metabolismo , Janus Quinasa 2/metabolismo , Terapia Molecular Dirigida , Metástasis de la Neoplasia , Fosfatidilinositol 3-Quinasa/metabolismo , Receptor ErbB-2/metabolismo , Transducción de Señal/efectos de los fármacos
7.
Breast Cancer Res ; 14(4): 318, 2012 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-22866917

RESUMEN

The Hippo pathway has emerged as a well-conserved kinase cascade controlling cell proliferation and survival and has recently gained much attention for its key activity as a tumor suppressor. In a study published in Cell, Cordenonsi and colleagues link TAZ, a downstream effector of the Hippo pathway, to attributes of putative breast cancer stem cells, epithelial-to-mesenchymal transition and cell polarity.


Asunto(s)
Neoplasias de la Mama/metabolismo , Células Madre Neoplásicas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Neoplasias de la Mama/patología , Activación Enzimática , Femenino , Vía de Señalización Hippo , Humanos
8.
Oncogene ; 41(39): 4459-4473, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36008466

RESUMEN

Plasticity delineates cancer subtypes with more or less favourable outcomes. In breast cancer, the subtype triple-negative lacks expression of major differentiation markers, e.g., estrogen receptor α (ERα), and its high cellular plasticity results in greater aggressiveness and poorer prognosis than other subtypes. Whether plasticity itself represents a potential vulnerability of cancer cells is not clear. However, we show here that cancer cell plasticity can be exploited to differentiate triple-negative breast cancer (TNBC). Using a high-throughput imaging-based reporter drug screen with 9 501 compounds, we have identified three polo-like kinase 1 (PLK1) inhibitors as major inducers of ERα protein expression and downstream activity in TNBC cells. PLK1 inhibition upregulates a cell differentiation program characterized by increased DNA damage, mitotic arrest, and ultimately cell death. Furthermore, cells surviving PLK1 inhibition have decreased tumorigenic potential, and targeting PLK1 in already established tumours reduces tumour growth both in cell line- and patient-derived xenograft models. In addition, the upregulation of genes upon PLK1 inhibition correlates with their expression in normal breast tissue and with better overall survival in breast cancer patients. Our results indicate that differentiation therapy based on PLK1 inhibition is a potential alternative strategy to treat TNBC.


Asunto(s)
Neoplasias de la Mama Triple Negativas , Mama/patología , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Proliferación Celular , Receptor alfa de Estrógeno , Humanos , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/metabolismo
9.
Br J Haematol ; 149(1): 55-64, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20096012

RESUMEN

Acute promyelocytic leukaemia (APL) patients are successfully treated with all-trans retinoic acid (ATRA). However, concurrent chemotherapy is still necessary and less toxic therapeutic approaches are needed. Earlier studies suggested that in haematopoietic neoplasms, the green tea polyphenol epigallocatechin-3-gallate (EGCG) induces cell death without adversely affecting healthy cells. We aimed at deciphering the molecular mechanism of EGCG-induced cell death in acute myeloid leukaemia (AML). A significant increase of death-associated protein kinase 2 (DAPK2) levels was found in AML cells upon EGCG treatment paralleled by increased cell death that was significantly reduced upon silencing of DAPK2. Moreover, combined ATRA and EGCG treatment resulted in cooperative DAPK2 induction and potentiated differentiation. EGCG toxicity of primary AML blasts correlated with 67 kDa laminin receptor (67LR) expression. Pretreatment of AML cells with ATRA, causing downregulation of 67LR, rendered these cells resistant to EGCG-mediated cell death. In summary, it was found that (i) DAPK2 is essential for EGCG-induced cell death in AML cells, (ii) ATRA and EGCG cotreatment significantly boosted neutrophil differentiation, and 67LR expression correlates with susceptibility of AML cells to EGCG. We thus suggest that EGCG, by selectively targeting leukaemic cells, may improve differentiation therapies for APL and chemotherapy for other AML subtypes.


Asunto(s)
Antineoplásicos/farmacología , Catequina/análogos & derivados , Leucemia Mieloide Aguda/patología , Neutrófilos/efectos de los fármacos , Tretinoina/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Proteínas Reguladoras de la Apoptosis/fisiología , Proteínas Quinasas Dependientes de Calcio-Calmodulina/fisiología , Catequina/farmacología , Muerte Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Proteínas Quinasas Asociadas a Muerte Celular , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos/métodos , Técnicas de Silenciamiento del Gen , Humanos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Células Tumorales Cultivadas , Regulación hacia Arriba/efectos de los fármacos
10.
ACS Chem Biol ; 14(10): 2215-2223, 2019 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-31553577

RESUMEN

Proteolysis targeting chimeras are bifunctional small molecules capable of recruiting a target protein of interest to an E3 ubiquitin ligase that facilitates target ubiquitination followed by proteasome-mediated degradation. The first molecules acting on this novel therapeutic paradigm have just entered clinical testing. Here, by using Bromodomain Containing 4 (BRD4) degraders engaging cereblon and Von Hippel-Lindau E3 ligases, we investigated key determinants of resistance to this new mode of action. A loss-of-function screen for genes required for BRD4 degradation revealed strong dependence on the E2 and E3 ubiquitin ligases as well as for members of the COP9 signalosome complex for both cereblon- and Von Hippel-Lindau-engaging BRD4 degraders. Cancer cell lines raised to resist BRD4 degraders manifested a degrader-specific mechanism of resistance, resulting from the loss of components of the ubiquitin proteasome system. In addition, degrader profiling in a cancer cell line panel revealed a differential pattern of activity of Von Hippel-Lindau- and cereblon-based degraders, highlighting the need for the identification of degradation-predictive biomarkers enabling effective patient stratification.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Azepinas/farmacología , Proteínas de Ciclo Celular/metabolismo , Resistencia a Medicamentos/efectos de los fármacos , Factores de Transcripción/metabolismo , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo , Proteínas de Ciclo Celular/química , Línea Celular Tumoral , Dipéptidos/farmacología , Células HEK293 , Humanos , Ftalimidas/farmacología , Prueba de Estudio Conceptual , Proteolisis , Factores de Transcripción/química , Ubiquitina-Proteína Ligasas/metabolismo
11.
J Leukoc Biol ; 81(6): 1599-608, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17347302

RESUMEN

The death-associated protein kinase 2 (DAPK2) belongs to a family of Ca(2+)/calmodulin-regulated serine/threonine kinases involved in apoptosis. During investigation of candidate genes operative in granulopoiesis, we identified DAPK2 as highly expressed. Subsequent investigations demonstrated particularly high DAPK2 expression in normal granulocytes compared with monocytes/macrophages and CD34(+) progenitor cells. Moreover, significantly increased DAPK2 mRNA levels were seen when cord blood CD34(+) cells were induced to differentiate toward neutrophils in tissue culture. In addition, all-trans retinoic acid (ATRA)-induced neutrophil differentiation of two leukemic cell lines, NB4 and U937, revealed significantly higher DAPK2 mRNA expression paralleled by protein induction. In contrast, during differentiation of CD34(+) and U937 cells toward monocytes/macrophages, DAPK2 mRNA levels remained low. In primary leukemia, low expression of DAPK2 was seen in acute myeloid leukemia samples, whereas chronic myeloid leukemia samples in chronic phase showed intermediate expression levels. Lentiviral vector-mediated expression of DAPK2 in NB4 cells enhanced, whereas small interfering RNA-mediated DAPK2 knockdown reduced ATRA-induced granulocytic differentiation, as evidenced by morphology and neutrophil stage-specific maturation genes, such as CD11b, G-CSF receptor, C/EBPepsilon, and lactoferrin. In summary, our findings implicate a role for DAPK2 in granulocyte maturation.


Asunto(s)
Proteínas Quinasas Dependientes de Calcio-Calmodulina/biosíntesis , Leucemia Mieloide/patología , Células Mieloides/citología , Mielopoyesis/fisiología , Neutrófilos/citología , Enfermedad Aguda , Antígenos CD34/metabolismo , Antígenos de Diferenciación/metabolismo , Proteínas Reguladoras de la Apoptosis , Diferenciación Celular , Línea Celular Tumoral , Enfermedad Crónica , Proteínas Quinasas Asociadas a Muerte Celular , Perfilación de la Expresión Génica , Granulocitos/citología , Granulocitos/fisiología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/fisiología , Humanos , Leucemia Mieloide/metabolismo , Células Mieloides/fisiología , Neutrófilos/fisiología , ARN Interferente Pequeño/biosíntesis , Tretinoina/farmacología , Regulación hacia Arriba
12.
Dis Aquat Organ ; 81(2): 119-25, 2008 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-18924376

RESUMEN

In recent years, numerous cases of morphological gonadal alterations in fish have been recorded throughout the world and across a wide range of species. In the whitefish Coregonus fatioi from the pre-alpine Lake Thun (Switzerland), the frequency of gonadal alterations is particularly high and the variety of alteration types large. Little is known about the proximal causes and the direct consequences of these morphological features on population persistence. In particular, the potential for the observed alterations to be the phenotypic expression of reduced genetic quality has not yet been addressed. In this study, we used offspring survival during embryogenesis as a proximate indicator of male genetic quality and tested whether the presence of gonadal alterations in males is an indicator of reduced quality. Embryos resulted from in vitro fertilizations of gametes from 126 males and females. We found no significant correlation between embryo survival and gonadal alteration in adults. Our findings suggest that in C. fatioi of Lake Thun, alterations in gonad morphology are not a phenotypic expression of variation in genetic quality.


Asunto(s)
Anomalías Congénitas/veterinaria , Enfermedades de los Peces/epidemiología , Gónadas/anomalías , Salmonidae/anomalías , Animales , Anomalías Congénitas/epidemiología , Trastornos del Desarrollo Sexual/patología , Trastornos del Desarrollo Sexual/veterinaria , Embrión no Mamífero , Femenino , Enfermedades de los Peces/patología , Agua Dulce , Masculino , Salmonidae/genética , Suiza/epidemiología
13.
Oxid Med Cell Longev ; 2018: 1482795, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29743969

RESUMEN

Autophagy is an intracellular degradation system that ensures a dynamic recycling of a variety of building blocks required for self-renewal, homeostasis, and cell survival under stress. We used primary acute myeloid leukemia (AML) samples and human AML cell lines to investigate the regulatory mechanisms of autophagy and its role in AML differentiation. We found a significantly lower expression of key autophagy- (ATG-) related genes in primary AML as compared to healthy granulocytes, an increased autophagic activity during all-trans retinoic acid- (ATRA-) induced neutrophil differentiation, and an impaired AML differentiation upon inhibition of ATG3, ATG4D, and ATG5. Supporting the notion of noncanonical autophagy, we found that ATRA-induced autophagy was Beclin1-independent compared to starvation- or arsenic trioxide- (ATO-) induced autophagy. Furthermore, we identified PU.1 as positive transcriptional regulator of ATG3, ATG4D, and ATG5. Low PU.1 expression in AML may account for low ATG gene expression in this disease. Low expression of the autophagy initiator ULK1 in AML can partially be attributed to high expression of the ULK1-targeting microRNA-106a. Our data clearly suggest that granulocytic AML differentiation relies on noncanonical autophagy pathways and that restoring autophagic activity might be beneficial in differentiation therapies.


Asunto(s)
Autofagia/genética , Diferenciación Celular/genética , Expresión Génica/genética , Leucemia Mieloide Aguda/genética , Línea Celular Tumoral , Humanos , Leucemia Mieloide Aguda/patología , Leucemia Mieloide Aguda/terapia , Fenotipo
14.
BMC Evol Biol ; 7: 207, 2007 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-17974038

RESUMEN

BACKGROUND: Males that are successful in intra-sexual competition are often assumed to be of superior quality. In the mating system of most salmonid species, intensive dominance fights are common and the winners monopolise most mates and sire most offspring. We drew a random sample of mature male brown trout (Salmo trutta) from two wild populations and determined their dominance hierarchy or traits linked to dominance. The fish were then stripped and their sperm was used for in vitro fertilisations in two full-factorial breeding designs. We recorded embryo viability until hatching in both experiments, and juvenile survival during 20 months after release into a natural streamlet in the second experiment. Since offspring of brown trout get only genes from their fathers, we used offspring survival as a quality measure to test (i) whether males differ in their genetic quality, and if so, (ii) whether dominance or traits linked to dominance reveal 'good genes'. RESULTS: We found significant additive genetic variance on embryo survival, i.e. males differed in their genetic quality. Older, heavier and larger males were more successful in intra-sexual selection. However, neither dominance nor dominance indicators like body length, weight or age were significantly linked to genetic quality measured as embryo or juvenile survival. CONCLUSION: We found no evidence that females can improve their offspring's genetic viability by mating with large and dominant males. If there still were advantages of mating with dominant males, they may be linked to non-genetic benefits or to genetic advantages that are context dependent and therefore possibly not revealed under our experimental conditions - even if we found significant additive genetic variation for embryo viability under such conditions.


Asunto(s)
Tamaño Corporal , Predominio Social , Trucha/genética , Envejecimiento , Animales , Femenino , Masculino , Reproducción , Trucha/anatomía & histología
15.
Mol Cell Biol ; 36(1): 132-43, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26483415

RESUMEN

Death-associated protein kinase 2 (DAPK2) is a Ca(2+)/calmodulin-dependent Ser/Thr kinase that possesses tumor-suppressive functions and regulates programmed cell death, autophagy, oxidative stress, hematopoiesis, and motility. As only few binding partners of DAPK2 have been determined, the molecular mechanisms governing these biological functions are largely unknown. We report the identification of 180 potential DAPK2 interaction partners by affinity purification-coupled mass spectrometry, 12 of which are known DAPK binding proteins. A small subset of established and potential binding proteins detected in this screen was further investigated by bimolecular fluorescence complementation (BiFC) assays, a method to visualize protein interactions in living cells. These experiments revealed that α-actinin-1 and 14-3-3-ß are novel DAPK2 binding partners. The interaction of DAPK2 with α-actinin-1 was localized at the plasma membrane, resulting in massive membrane blebbing and reduced cellular motility, whereas the interaction of DAPK2 with 14-3-3-ß was localized to the cytoplasm, with no impact on blebbing, motility, or viability. Our results therefore suggest that DAPK2 effector functions are influenced by the protein's subcellular localization and highlight the utility of combining mass spectrometry screening with bimolecular fluorescence complementation to identify and characterize novel protein-protein interactions.


Asunto(s)
Apoptosis/fisiología , Proteínas Quinasas Asociadas a Muerte Celular/metabolismo , Fluorescencia , Proteómica , Autofagia/fisiología , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Células Cultivadas , Humanos , Unión Proteica/fisiología , Proteómica/métodos
16.
Exp Hematol ; 43(9): 781-93.e2, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25986473

RESUMEN

Acute myeloid leukemia (AML) is characterized by the accumulation of immature blood cell precursors in the bone marrow. Pharmacologically overcoming the differentiation block in this condition is an attractive therapeutic avenue, which has achieved success only in a subtype of AML, acute promyelocytic leukemia (APL). Attempts to emulate this success in other AML subtypes have thus far been unsuccessful. Autophagy is a conserved protein degradation pathway with important roles in mammalian cell differentiation, particularly within the hematopoietic system. In the study described here, we investigated the functional importance of autophagy in APL cell differentiation. We found that autophagy is increased during all-trans-retinoic acid (ATRA)-induced granulocytic differentiation of the APL cell line NB4 and that this is associated with increased expression of LC3II and GATE-16 proteins involved in autophagosome formation. Autophagy inhibition, using either drugs (chloroquine/3-methyladenine) or short-hairpin RNA targeting the essential autophagy gene ATG7, attenuates myeloid differentiation. Importantly, we found that enhancing autophagy promotes ATRA-induced granulocytic differentiation of an ATRA-resistant derivative of the non-APL AML HL60 cell line (HL60-Diff-R). These data support the development of strategies to stimulate autophagy as a novel approach to promote differentiation in AML.


Asunto(s)
Antineoplásicos/farmacología , Autofagia/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Leucemia Promielocítica Aguda/tratamiento farmacológico , Tretinoina/farmacología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adenina/análogos & derivados , Adenina/farmacología , Antirreumáticos/farmacología , Proteína 7 Relacionada con la Autofagia , Familia de las Proteínas 8 Relacionadas con la Autofagia , Cloroquina/farmacología , Granulocitos/metabolismo , Granulocitos/patología , Células HL-60 , Humanos , Leucemia Promielocítica Aguda/genética , Leucemia Promielocítica Aguda/metabolismo , Leucemia Promielocítica Aguda/patología , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Enzimas Activadoras de Ubiquitina/antagonistas & inhibidores , Enzimas Activadoras de Ubiquitina/genética , Enzimas Activadoras de Ubiquitina/metabolismo
17.
J Leukoc Biol ; 95(1): 83-93, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24038216

RESUMEN

DAPK2 is a proapoptotic protein that is mostly expressed in the hematopoietic tissue. A detailed DAPK2 expression analysis in two large AML patient cohorts revealed particularly low DAPK2 mRNA levels in APL. DAPK2 levels were restored in APL patients undergoing ATRA therapy. PML-RARA is the predominant lesion in APL causing transcriptional repression of genes important for neutrophil differentiation. We found binding of PML-RARA and PU.1, a myeloid master regulator, to RARA and PU.1 binding sites in the DAPK2 promoter. Ectopic expression of PML-RARA in non-APL, as well as knocking down PU.1 in APL cells, resulted in a significant reduction of DAPK2 expression. Restoring DAPK2 expression in PU.1 knockdown APL cells partially rescued neutrophil differentiation, thereby identifying DAPK2 as a relevant PU.1 downstream effector. Moreover, low DAPK2 expression is also associated with C/EBPα-mutated AML patients, and we found C/EBPα-dependent regulation of DAPK2 during APL differentiation. In conclusion, we identified first inhibitory mechanisms responsible for the low DAPK2 expression in particular AML subtypes, and the regulation of DAPK2 by two myeloid transcription factors underlines its importance in neutrophil development.


Asunto(s)
Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Diferenciación Celular/genética , Proteínas Quinasas Asociadas a Muerte Celular/genética , Regulación de la Expresión Génica , Granulocitos/citología , Granulocitos/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Transactivadores/metabolismo , Línea Celular Tumoral , Proteínas Quinasas Asociadas a Muerte Celular/metabolismo , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Promielocítica Aguda/genética , Leucemia Promielocítica Aguda/metabolismo , Neutrófilos/citología , Neutrófilos/metabolismo , Transcripción Genética
18.
Cancer Cell ; 22(6): 796-811, 2012 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-23238015

RESUMEN

Hyperactive PI3K/mTOR signaling is prevalent in human malignancies and its inhibition has potent antitumor consequences. Unfortunately, single-agent targeted cancer therapy is usually short-lived. We have discovered a JAK2/STAT5-evoked positive feedback loop that dampens the efficacy of PI3K/mTOR inhibition. Mechanistically, PI3K/mTOR inhibition increased IRS1-dependent activation of JAK2/STAT5 and secretion of IL-8 in several cell lines and primary breast tumors. Genetic or pharmacological inhibition of JAK2 abrogated this feedback loop and combined PI3K/mTOR and JAK2 inhibition synergistically reduced cancer cell number and tumor growth, decreased tumor seeding and metastasis, and also increased overall survival of the animals. Our results provide a rationale for combined targeting of the PI3K/mTOR and JAK2/STAT5 pathways in triple-negative breast cancer, a particularly aggressive and currently incurable disease.


Asunto(s)
Neoplasias de la Mama/metabolismo , Janus Quinasa 2/antagonistas & inhibidores , Metástasis de la Neoplasia/tratamiento farmacológico , Inhibidores de las Quinasa Fosfoinosítidos-3 , Factor de Transcripción STAT5/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Animales , Neoplasias de la Mama/tratamiento farmacológico , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Femenino , Humanos , Proteínas Sustrato del Receptor de Insulina/metabolismo , Interleucina-8/metabolismo , Janus Quinasa 2/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Ratones SCID , Fosfatidilinositol 3-Quinasas/metabolismo , Receptores de Interleucina-8A/metabolismo , Factor de Transcripción STAT5/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
19.
Mol Cancer Res ; 9(10): 1377-84, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21849469

RESUMEN

Protein-tyrosine phosphatase 1B (PTP1B), a well-established metabolic regulator, plays an important role in breast cancer. Using whole-body PTP1B knockout mice, recent studies have shown that PTP1B ablation delays HER2/Neu-induced mammary cancer. Whether PTP1B plays a cell-autonomous or a noncell-autonomous role in HER2/Neu-evoked tumorigenesis and whether it is involved in tumor maintenance was unknown. We generated mice expressing HER2/Neu and lacking PTP1B specifically in the mammary epithelium. We found that mammary-specific deletion of PTP1B delays the onset of HER2/Neu-evoked mammary tumors, establishing a cell autonomous role for PTP1B in such neoplasms. We also deleted PTP1B in established mouse mammary tumors or depleted PTP1B in human breast cancer cell lines grown as xenografts. PTP1B inhibition did not affect tumor growth in either model showing that neither epithelial nor stromal PTP1B is necessary for tumor maintenance. Taken together, our data show that despite the PTP1B contribution to tumor onset, it is not essential for tumor maintenance. This suggests that PTP1B inhibition could be effective in breast tumor prevention.


Asunto(s)
Neoplasias Mamarias Experimentales/enzimología , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Receptor ErbB-2/metabolismo , Animales , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Femenino , Humanos , Inmunohistoquímica , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Noqueados , Ratones SCID , Ratones Transgénicos , Proteína Tirosina Fosfatasa no Receptora Tipo 1/deficiencia , Proteína Tirosina Fosfatasa no Receptora Tipo 1/genética , Transducción de Señal
20.
Mol Cancer Res ; 7(6): 916-22, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19491197

RESUMEN

The Hypermethylated in Cancer 1 (HIC1) gene encodes a zinc finger transcriptional repressor that cooperates with p53 to suppress cancer development. We and others recently showed that HIC1 is a transcriptional target of p53. To identify additional transcriptional regulators of HIC1, we screened a set of transcription factors for regulation of a human HIC1 promoter reporter. We found that E2F1 strongly activates the full-length HIC1 promoter reporter. Promoter deletions and mutations identified two E2F responsive elements in the HIC1 core promoter region. Moreover, in vivo binding of E2F1 to the HIC1 promoter was shown by chromatin immunoprecipitation assays in human TIG3 fibroblasts expressing tamoxifen-activated E2F1. In agreement, activation of E2F1 in TIG3-E2F1 cells markedly increased HIC1 expression. Interestingly, expression of E2F1 in the p53(-/-) hepatocellular carcinoma cell line Hep3B led to an increase of endogenous HIC1 mRNA, although bisulfite genomic sequencing of the HIC1 promoter revealed that the region bearing the two E2F1 binding sites is hypermethylated. In addition, endogenous E2F1 induced by etoposide treatment bound to the HIC1 promoter. Moreover, inhibition of E2F1 strongly reduced the expression of etoposide-induced HIC1. In conclusion, we identified HIC1 as novel E2F1 transcriptional target in DNA damage responses.


Asunto(s)
Factor de Transcripción E2F1/genética , Factores de Transcripción de Tipo Kruppel/genética , Secuencia de Bases , Sitios de Unión , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Línea Celular Tumoral , Metilación de ADN , Factor de Transcripción E2F1/metabolismo , Etopósido/farmacología , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Factores de Transcripción de Tipo Kruppel/biosíntesis , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Alineación de Secuencia , Transcripción Genética , Regulación hacia Arriba/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA