Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 18(3): 293-302, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28092373

RESUMEN

The aggregation of hypertrophic macrophages constitutes the basis of all granulomatous diseases, such as tuberculosis or sarcoidosis, and is decisive for disease pathogenesis. However, macrophage-intrinsic pathways driving granuloma initiation and maintenance remain elusive. We found that activation of the metabolic checkpoint kinase mTORC1 in macrophages by deletion of the gene encoding tuberous sclerosis 2 (Tsc2) was sufficient to induce hypertrophy and proliferation, resulting in excessive granuloma formation in vivo. TSC2-deficient macrophages formed mTORC1-dependent granulomatous structures in vitro and showed constitutive proliferation that was mediated by the neo-expression of cyclin-dependent kinase 4 (CDK4). Moreover, mTORC1 promoted metabolic reprogramming via CDK4 toward increased glycolysis while simultaneously inhibiting NF-κB signaling and apoptosis. Inhibition of mTORC1 induced apoptosis and completely resolved granulomas in myeloid TSC2-deficient mice. In human sarcoidosis patients, mTORC1 activation, macrophage proliferation and glycolysis were identified as hallmarks that correlated with clinical disease progression. Collectively, TSC2 maintains macrophage quiescence and prevents mTORC1-dependent granulomatous disease with clinical implications for sarcoidosis.


Asunto(s)
Granuloma/inmunología , Macrófagos/inmunología , Complejos Multiproteicos/metabolismo , Sarcoidosis/inmunología , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Línea Celular , Quinasa 4 Dependiente de la Ciclina/metabolismo , Progresión de la Enfermedad , Granuloma/tratamiento farmacológico , Humanos , Macrófagos/efectos de los fármacos , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Interferente Pequeño/genética , Sarcoidosis/tratamiento farmacológico , Transducción de Señal , Proteína 2 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/genética
2.
Cell ; 147(2): 306-19, 2011 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-22000011

RESUMEN

Chronic myelogenous leukemia (CML) is caused by the constitutively active tyrosine kinase Bcr-Abl and treated with the tyrosine kinase inhibitor (TKI) imatinib. However, emerging TKI resistance prevents complete cure. Therefore, alternative strategies targeting regulatory modules of Bcr-Abl in addition to the kinase active site are strongly desirable. Here, we show that an intramolecular interaction between the SH2 and kinase domains in Bcr-Abl is both necessary and sufficient for high catalytic activity of the enzyme. Disruption of this interface led to inhibition of downstream events critical for CML signaling and, importantly, completely abolished leukemia formation in mice. Furthermore, disruption of the SH2-kinase interface increased sensitivity of imatinib-resistant Bcr-Abl mutants to TKI inhibition. An engineered Abl SH2-binding fibronectin type III monobody inhibited Bcr-Abl kinase activity both in vitro and in primary CML cells, where it induced apoptosis. This work validates the SH2-kinase interface as an allosteric target for therapeutic intervention.


Asunto(s)
Proteínas de Fusión bcr-abl/antagonistas & inhibidores , Proteínas de Fusión bcr-abl/química , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/enzimología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/química , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Benzamidas , Células Cultivadas , Proteínas de Fusión bcr-abl/metabolismo , Humanos , Mesilato de Imatinib , Isoleucina/metabolismo , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Piperazinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas/metabolismo , Pirimidinas/farmacología , Transducción de Señal , Dominios Homologos src
3.
Arch Toxicol ; 90(9): 2173-2186, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27325309

RESUMEN

Since the discovery of human embryonic stem cells (hESC) and human-induced pluripotent stem cells (hiPSC), great hopes were held for their therapeutic application including disease modeling, drug discovery screenings, toxicological screenings and regenerative therapy. hESC and hiPSC have the advantage of indefinite self-renewal, thereby generating an inexhaustible pool of cells with, e.g., specific genotype for developing putative treatments; they can differentiate into derivatives of all three germ layers enabling autologous transplantation, and via donor-selection they can express various genotypes of interest for better disease modeling. Furthermore, drug screenings and toxicological screenings in hESC and hiPSC are more pertinent to identify drugs or chemical compounds that are harmful for human, than a mouse model could predict. Despite continuing research in the wide field of therapeutic applications, further understanding of the underlying basic mechanisms of stem cell function is necessary. Here, we summarize current knowledge concerning pluripotency, self-renewal, apoptosis, motility, epithelial-to-mesenchymal transition and differentiation of pluripotent stem cells.


Asunto(s)
Diferenciación Celular , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Pruebas de Toxicidad/métodos , Investigación Biomédica Traslacional/métodos , Alternativas a las Pruebas en Animales , Apoptosis/efectos de los fármacos , Bioensayo , Ciclo Celular/efectos de los fármacos , Linaje de la Célula , Movimiento Celular/efectos de los fármacos , Autorrenovación de las Células/efectos de los fármacos , Transformación Celular Neoplásica/inducido químicamente , Células Cultivadas , Transición Epitelial-Mesenquimal/efectos de los fármacos , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Fenotipo , Medición de Riesgo
4.
Eur J Clin Invest ; 45(4): 415-22, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25646864

RESUMEN

BACKGROUND: The ability of cells to travel long distances in order to form tissues and organs is inherently connected to embryogenesis. The process in which epithelial-like embryonic cells become motile and invasive is termed 'epithelial-to-mesenchymal transition' (EMT), while the reversion of this programme--yielding differentiated cells and organs--is called 'mesenchymal-to-epithelial transition' (MET). DESIGN: Here, we review the processes of EMT and MET in development and cancer and combine them with knowledge from pluripotent stem cell research. RESULTS: Research has shown that these processes are activated in many cancers leading to dissemination of cancer cells throughout the body and formation of metastasis. While the regulation of EMT during cancer progression has been extensively studied for decades, many fundamental processes that govern normal development are only poorly understood. Recent discoveries, such as reprogramming to pluripotent stem cells and identification of ground and primed states of pluripotent stem cells, have redirected much attention to EMT and MET. CONCLUSION: Findings from pluripotent stem cell research and EMT/MET should be combined in order to design future strategies aimed to improve our understanding of cancer progression and to help develop novel anticancer strategies.


Asunto(s)
Transición Epitelial-Mesenquimal/fisiología , Neoplasias/fisiopatología , Células Madre Pluripotentes/fisiología , Diferenciación Celular , Desarrollo Embrionario , Humanos
5.
Haematologica ; 99(6): 1006-15, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24561791

RESUMEN

Considerable effort has been expended to identify genes that account for myeloid lineage commitment and development. However, currently available non-invasive mouse models utilize myeloid-specific reporters that are significantly expressed in hematopoietic stem cells as well as lymphoid compartments. Here, we describe a myeloid-specific marker that is not shared by any other lineage. We show that lactotransferrin mRNA is expressed by Gr-1(+)/CD11b(+) cells in the bone marrow, as opposed to hematopoietic stem cells or any peripheral cell population. To follow the progeny of lactotransferrin-expressing bone marrow cells, we generated a mouse model in which a reporter gene is irreversibly activated from the lactotransferrin-promoter. We found that lactotransferrin-reporter labels a majority of neutrophils, monocytes, macrophages and distinct subtypes of dendritic cells, while excluding T, B, natural killer cells, interferon-producing killer dendritic cells, plasmacytoid dendritic cells, erythrocytes and eosinophils. Lactotransferrin-reporter(-) bone marrow cells retain lymphoid, erythroid and long-term repopulating potential, while lactotransferrin-reporter(+) bone marrow cells confer only myeloid, but not lymphoid potential. We conclude that lactotransferrin represents a late stage differentiation marker of neutrophils, macrophages and distinct subtypes of dendritic cells.


Asunto(s)
Células Dendríticas/metabolismo , Lactoferrina/genética , Macrófagos/metabolismo , Monocitos/metabolismo , Neutrófilos/metabolismo , Animales , Antígeno CD11b/metabolismo , Rastreo Celular , Células Eritroides/metabolismo , Expresión Génica , Orden Génico , Genes Reporteros , Vectores Genéticos/genética , Lactoferrina/metabolismo , Linfocitos/metabolismo , Ratones , Ratones Transgénicos , Células Mieloides/metabolismo , Especificidad de Órganos/genética , Regiones Promotoras Genéticas , ARN Mensajero/genética , Receptores de Quimiocina/metabolismo
6.
Cancer Cell ; 10(1): 77-87, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16843267

RESUMEN

The tumor suppressor STAT1 is considered a key regulator of the surveillance of developing tumors. Here, we describe an unexpected tumor-promoting role for STAT1 in leukemia. STAT1(-/-) mice are partially protected from leukemia development, and STAT1(-/-) tumor cells induce leukemia in RAG2(-/-) and immunocompetent mice with increased latency. The low MHC class I protein levels of STAT1(-/-) tumor cells enable efficient NK cell lysis and account for the enhanced tumor clearance. Strikingly, STAT1(-/-) tumor cells acquire increased MHC class I expression upon leukemia progression. These findings define STAT1 as a tumor promoter in leukemia development. Furthermore, we describe the upregulation of MHC class I expression as a general mechanism that allows for the escape of hematopoietic malignancies from immune surveillance.


Asunto(s)
Leucemia Experimental/patología , Factor de Transcripción STAT1/fisiología , Animales , Linfocitos B/metabolismo , Linfocitos B/patología , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular/genética , Transformación Celular Neoplásica/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Progresión de la Enfermedad , Genotipo , Antígenos de Histocompatibilidad Clase I/inmunología , Antígenos de Histocompatibilidad Clase I/metabolismo , Interferón gamma/genética , Interferón gamma/metabolismo , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Leucemia Experimental/genética , Leucemia Experimental/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Oncogénicas v-abl/genética , Proteínas Oncogénicas v-abl/metabolismo , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Fenotipo , Factor de Transcripción STAT1/deficiencia , Factor de Transcripción STAT1/genética , Células Madre/metabolismo , Células Madre/patología , Análisis de Supervivencia
7.
Blood ; 118(17): 4635-45, 2011 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-21878673

RESUMEN

In Eµ-myc transgenic animals lymphoma formation requires additional genetic alterations, which frequently comprise loss of p53 or overexpression of BCL-2. We describe that the nature of the "second hit" affects the ability of the immune system to contain lymphoma development. Tumors with disrupted p53 signaling killed the host more rapidly than BCL-2 overexpressing ones. Relaxing immunologic control, using Tyk2(-/-) mice or by Ab-mediated depletion of CD8(+) T or natural killer (NK) cells accelerated formation of BCL-2-overexpressing lymphomas but not of those lacking p53. Most strikingly, enforced expression of BCL-2 prolonged disease latency in the absence of p53, whereas blocking p53 function in BCL-2-overexpressing tumors failed to accelerate disease. This shows that blocking apoptosis in p53-deficient cells by enforcing BCL-2 expression can mitigate disease progression increasing the "immunologic visibility." In vitro cytotoxicity assays confirmed that high expression of BCL-2 protein facilitates NK and T cell-mediated killing. Moreover, we found that high BCL-2 expression is accompanied by significantly increased levels of the NKG2D ligand MULT1, which may account for the enhanced killing. Our findings provide first evidence that the nature of the second hit affects tumor immunosurveillance in c-MYC-driven lymphomas and define a potential shortcoming of antitumor therapies targeting BCL-2.


Asunto(s)
Epistasis Genética/inmunología , Genes myc/fisiología , Vigilancia Inmunológica/genética , Linfoma/genética , Mutación/fisiología , Animales , Transformación Celular Neoplásica/genética , Células Cultivadas , Progresión de la Enfermedad , Epistasis Genética/fisiología , Genes bcl-2/fisiología , Genes p53/fisiología , Linfoma/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , TYK2 Quinasa/genética , Escape del Tumor/genética
8.
Blood ; 116(9): 1548-58, 2010 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-20508164

RESUMEN

Stat5 transcription factors are essential gene regulators promoting proliferation, survival, and differentiation of all hematopoietic cell types. Mutations or fusions of oncogenic tyrosine kinases often result in constitutive Stat5 activation. We have modeled persistent Stat5 activity by using an oncogenic Stat5a variant (cS5). To analyze the hitherto unrecognized role of Stat5 serine phosphorylation in this context, we have generated cS5 constructs with mutated C-terminal serines 725 and 779, either alone or in combination. Genetic complementation assays in primary Stat5(null/null) mast cells and Stat5(DeltaN) T cells demonstrated reconstitution of proliferation with these mutants. Similarly, an in vivo reconstitution experiment of transduced Stat5(null/null) fetal liver cells transplanted into irradiated wild-type recipients revealed that these mutants exhibit biologic activity in lineage differentiation. By contrast, the leukemogenic potential of cS5 in bone marrow transplants decreased dramatically in cS5 single-serine mutants or was completely absent upon loss of both serine phosphorylation sites. Our data suggest that Stat5a serine phosphorylation is a prerequisite for cS5-mediated leukemogenesis. Hence, interference with Stat5a serine phosphorylation might provide a new therapeutic option for leukemia and myeloid dysplasias without affecting major functions of Stat5 in normal hematopoiesis.


Asunto(s)
Transformación Celular Neoplásica , Hematopoyesis/fisiología , Leucemia/patología , Factor de Transcripción STAT5/metabolismo , Serina/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Adulto , Anciano , Animales , Western Blotting , Trasplante de Médula Ósea , Linaje de la Célula , Proliferación Celular , Células Cultivadas , Femenino , Feto , Citometría de Flujo , Humanos , Técnicas para Inmunoenzimas , Leucemia/genética , Leucemia/metabolismo , Trasplante de Hígado , Masculino , Mastocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Fosforilación , Células Precursoras de Linfocitos B/metabolismo , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT5/genética , Serina/genética , Linfocitos T/metabolismo , Proteínas Supresoras de Tumor/genética
9.
Oncogene ; 40(6): 1091-1105, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33323974

RESUMEN

Metastatic melanoma is hallmarked by its ability of phenotype switching to more slowly proliferating, but highly invasive cells. Here, we tested the impact of signal transducer and activator of transcription 3 (STAT3) on melanoma progression in association with melanocyte inducing transcription factor (MITF) expression levels. We established a mouse melanoma model for deleting Stat3 in melanocytes with specific expression of human hyperactive NRASQ61K in an Ink4a-deficient background, two frequent driver mutations in human melanoma. Mice devoid of Stat3 showed early disease onset with higher proliferation in primary tumors, but displayed significantly diminished lung, brain, and liver metastases. Whole-genome expression profiling of tumor-derived cells also showed a reduced invasion phenotype, which was further corroborated by 3D melanoma model analysis. Notably, loss or knockdown of STAT3 in mouse or human cells resulted in the upregulation of MITF and induction of cell proliferation. Mechanistically we show that STAT3-induced CAAT Box Enhancer Binding Protein (CEBP) expression was sufficient to suppress MITF transcription. Epigenetic analysis by ATAC-seq confirmed that CEBPa/b binding to the MITF enhancer region silenced the MITF locus. Finally, by classification of patient-derived melanoma samples, we show that STAT3 and MITF act antagonistically and hence contribute differentially to melanoma progression. We conclude that STAT3 is a driver of the metastatic process in melanoma and able to antagonize MITF via direct induction of CEBP family member transcription.


Asunto(s)
Proteína beta Potenciadora de Unión a CCAAT/genética , Melanoma/genética , Factor de Transcripción Asociado a Microftalmía/genética , Factor de Transcripción STAT3/genética , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Melanocitos/efectos de los fármacos , Melanoma/patología , Ratones , Metástasis de la Neoplasia , Transducción de Señal/efectos de los fármacos
10.
Blood ; 112(9): 3878-88, 2008 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-18694996

RESUMEN

Erythropoiesis strictly depends on signal transduction through the erythropoietin receptor (EpoR)-Janus kinase 2 (Jak2)-signal transducer and activator of transcription 5 (Stat5) axis, regulating proliferation, differentiation, and survival. The exact role of the transcription factor Stat5 in erythropoiesis remained puzzling, however, since the first Stat5-deficient mice carried a hypomorphic Stat5 allele, impeding full phenotypical analysis. Using mice completely lacking Stat5--displaying early lethality--we demonstrate that these animals suffer from microcytic anemia due to reduced expression of the antiapoptotic proteins Bcl-x(L) and Mcl-1 followed by enhanced apoptosis. Moreover, transferrin receptor-1 (TfR-1) cell surface levels on erythroid cells were decreased more than 2-fold on erythroid cells of Stat5(-/-) animals. This reduction could be attributed to reduced transcription of TfR-1 mRNA and iron regulatory protein 2 (IRP-2), the major translational regulator of TfR-1 mRNA stability in erythroid cells. Both genes were demonstrated to be direct transcriptional targets of Stat5. This establishes an unexpected mechanistic link between EpoR/Jak/Stat signaling and iron metabolism, processes absolutely essential for erythropoiesis and life.


Asunto(s)
Células Eritroides/metabolismo , Proteína 2 Reguladora de Hierro/metabolismo , Hierro/metabolismo , Receptores de Transferrina/metabolismo , Factor de Transcripción STAT5/metabolismo , Anemia Ferropénica/genética , Anemia Ferropénica/metabolismo , Anemia Ferropénica/patología , Animales , Apoptosis , Transporte Biológico Activo , Pérdida del Embrión , Células Eritroides/patología , Femenino , Deficiencias de Hierro , Hígado/embriología , Hígado/metabolismo , Hígado/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Embarazo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factor de Transcripción STAT5/deficiencia , Factor de Transcripción STAT5/genética
11.
Blood ; 111(9): 4511-22, 2008 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-18239084

RESUMEN

Erythropoiesis requires erythropoietin (Epo) and stem cell factor (SCF) signaling via their receptors EpoR and c-Kit. EpoR, like many other receptors involved in hematopoiesis, acts via the kinase Jak2. Deletion of EpoR or Janus kinase 2 (Jak2) causes embryonic lethality as a result of defective erythropoiesis. The contribution of distinct EpoR/Jak2-induced signaling pathways (mitogen-activated protein kinase, phosphatidylinositol 3-kinase, signal transducer and activator of transcription 5 [Stat5]) to functional erythropoiesis is incompletely understood. Here we demonstrate that expression of a constitutively activated Stat5a mutant (cS5) was sufficient to relieve the proliferation defect of Jak2(-/-) and EpoR(-/-) cells in an Epo-independent manner. In addition, tamoxifen-induced DNA binding of a Stat5a-estrogen receptor (ER)* fusion construct enabled erythropoiesis in the absence of Epo. Furthermore, c-Kit was able to enhance signaling through the Jak2-Stat5 axis, particularly in lymphoid and myeloid progenitors. Although abundance of hematopoietic stem cells was 2.5-fold reduced in Jak2(-/-) fetal livers, transplantation of Jak2(-/-)-cS5 fetal liver cells into irradiated mice gave rise to mature erythroid and myeloid cells of donor origin up to 6 months after transplantation. Cytokine- and c-Kit pathways do not function independently of each other in hematopoiesis but cooperate to attain full Jak2/Stat5 activation. In conclusion, activated Stat5 is a critical downstream effector of Jak2 in erythropoiesis/myelopoiesis, and Jak2 functionally links cytokine- with c-Kit-receptor tyrosine kinase signaling.


Asunto(s)
Eritropoyesis , Janus Quinasa 2 , Receptores de Eritropoyetina , Factor de Transcripción STAT5/metabolismo , Animales , Células Cultivadas , Citocinas/metabolismo , Ratones , Ratones Noqueados , Mielopoyesis , Proteínas Proto-Oncogénicas c-kit/metabolismo
12.
Nat Commun ; 11(1): 4166, 2020 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-32820173

RESUMEN

T cells engineered to express chimeric antigen receptors (CAR-T cells) have shown impressive clinical efficacy in the treatment of B cell malignancies. However, the development of CAR-T cell therapies for solid tumors is hampered by the lack of truly tumor-specific antigens and poor control over T cell activity. Here we present an avidity-controlled CAR (AvidCAR) platform with inducible and logic control functions. The key is the combination of (i) an improved CAR design which enables controlled CAR dimerization and (ii) a significant reduction of antigen-binding affinities to introduce dependence on bivalent interaction, i.e. avidity. The potential and versatility of the AvidCAR platform is exemplified by designing ON-switch CARs, which can be regulated with a clinically applied drug, and AND-gate CARs specifically recognizing combinations of two antigens. Thus, we expect that AvidCARs will be a highly valuable platform for the development of controllable CAR therapies with improved tumor specificity.


Asunto(s)
Inmunoterapia Adoptiva/métodos , Receptores de Antígenos de Linfocitos T/inmunología , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T/inmunología , Animales , Antígenos de Neoplasias/inmunología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Células Cultivadas , Citocinas/inmunología , Citocinas/metabolismo , Citotoxicidad Inmunológica/inmunología , Humanos , Activación de Linfocitos/inmunología , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Neoplasias/inmunología , Neoplasias/patología , Neoplasias/terapia , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/metabolismo , Linfocitos T/metabolismo
13.
Sci Rep ; 9(1): 1897, 2019 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-30760778

RESUMEN

Genetic and biochemical screening approaches often fail to identify functionally relevant pathway networks because many signaling proteins contribute to multiple gene ontology pathways. We developed a DRUGPATH-approach to predict pathway-interactomes from high-content drug screen data. DRUGPATH is based upon combining z-scores of effective inhibitors with their corresponding and validated targets. We test DRUGPATH by comparing homeostatic pathways in human embryonic stem cells (hESCs), human induced pluripotent stem cells (hiPSCs) and human amniotic fluid stem cells (hAFSCs). We show that hAFSCs utilize distinct interactomes compared to hESCs/hiPSCs and that pathways orchestrating cell cycle and apoptosis are strongly interconnected, while pathways regulating survival and size are not. Interestingly, hESCs/hiPSCs regulate their size by growing exact additional sizes during each cell cycle. Chemical and genetic perturbation studies show that this "adder-model" is dependent on the DNA-damage pathway. In the future, the DRUGPATH-approach may help to predict novel pathway interactomes from high-content drug screens.


Asunto(s)
Tamaño de la Célula/efectos de los fármacos , Biología Computacional/métodos , Daño del ADN , Inhibidores Enzimáticos/farmacología , Apoptosis/efectos de los fármacos , Proteínas de la Ataxia Telangiectasia Mutada/antagonistas & inhibidores , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Dimetilsulfóxido/farmacología , Células Madre Embrionarias Humanas , Humanos , Indazoles/farmacología , Células Madre Pluripotentes Inducidas , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Sulfonamidas/farmacología
14.
Front Biosci ; 13: 6237-54, 2008 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-18508657

RESUMEN

Stat5 proteins modulate gene transcription upon cytokine- and growth factor action. Stat5a and Stat5b proteins alone are weak activators of transcription. They can modify chromatin organization through oligomerization and they act predominantly in co-operation and interaction with other proteins. The conservative view of exclusively nuclear functions of Stat5 was challenged by the observation of additional Stat5 effects in the cytoplasm, resulting in activation of the PI3K-Akt pathway. We summarize biological consequences of mutations in conserved domains of Stat5 or of deletions in the N- or C-terminal domains with impact on target gene transcription. Formation of higher-order oligomers is dramatically changed upon amino- or carboxyterminal deletions in Stat5 proteins. Mutations in or deletion of the Stat5 N-terminus leads to diminished leukemogenic potential of oncogenic Stat5, probably due to the inability to form Stat5 tetramers. The Stat5 N-terminal domain prevents persistent activation and can act as a DNA-docking platform for the glucocorticoid receptor (GR). The corresponding protocols should facilitate follow-up studies on Stat5 proteins and their contribution to normal physiological versus pathological processes through differential chromatin binding.


Asunto(s)
Cromatina/fisiología , Factor de Transcripción STAT5/fisiología , Animales , Enfermedades Autoinmunes/fisiopatología , ADN/genética , ADN/metabolismo , Humanos , Inflamación/fisiopatología , Ratones , Ratones Noqueados , Modelos Animales , Trastornos Mieloproliferativos/fisiopatología , Neoplasias/fisiopatología , Isoformas de Proteínas/fisiología , Factor de Transcripción STAT5/deficiencia , Factor de Transcripción STAT5/genética
15.
J Clin Invest ; 114(11): 1650-8, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15578097

RESUMEN

Aberrant activation of the JAK-STAT pathway has been implicated in tumor formation; for example, constitutive activation of JAK2 kinase or the enforced expression of STAT5 induces leukemia in mice. We show here that the Janus kinase TYK2 serves an opposite function. Mice deficient in TYK2 developed Abelson-induced B lymphoid leukemia/lymphoma as well as TEL-JAK2-induced T lymphoid leukemia with a higher incidence and shortened latency compared with WT controls. The cell-autonomous properties of Abelson murine leukemia virus-transformed (A-MuLV-transformed) TYK2(-/-) cells were unaltered, but the high susceptibility of TYK2(-/-) mice resulted from an impaired tumor surveillance, and accordingly, TYK2(-/-) A-MuLV-induced lymphomas were easily rejected after transplantation into WT hosts. The increased rate of leukemia/lymphoma formation was linked to a decreased in vitro cytotoxic capacity of TYK2(-/-) NK and NKT cells toward tumor-derived cells. RAG2/TYK2 double-knockout mice succumbed to A-MuLV-induced leukemia/lymphoma faster than RAG2(-/-)TYK2(+/-) mice. This defines NK cells as key players in tumor surveillance in Abelson-induced malignancies. Our observations provide compelling evidence that TYK2 is an important regulator of lymphoid tumor surveillance.


Asunto(s)
Leucemia de Células B/inmunología , Leucemia Experimental/inmunología , Proteínas Tirosina Quinasas/metabolismo , Virus de la Leucemia Murina de Abelson/genética , Virus de la Leucemia Murina de Abelson/metabolismo , Animales , Animales Recién Nacidos , Transformación Celular Neoplásica , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Humanos , Leucemia de Células B/patología , Leucemia Experimental/patología , Leucemia de Células T/inmunología , Leucemia de Células T/patología , Hígado/citología , Hígado/patología , Ratones , Ratones Noqueados , Ratones Desnudos , Trasplante de Neoplasias , Proteínas Nucleares , Proteínas Tirosina Quinasas/genética , Bazo/citología , Bazo/patología , Tasa de Supervivencia , TYK2 Quinasa
16.
J Invest Dermatol ; 124(6): 1300-7, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15955107

RESUMEN

Clusterin has recently been shown to act as an antiapoptotic protein that confers drug-resistance in models of epithelial tumors. The aim of our work was to provide an insight into a possible role of clusterin in the regulation of drug-resistance in melanoma. In tissue samples, clusterin expression was low in nevi, but high in primary melanoma and melanoma metastases. Clusterin was also strongly expressed in melanoma cell lines, but was barely detectable in cultured melanocytes. To elucidate a possible role of clusterin in drug-resistance of melanoma, clusterin expression was regulated by either plasmid-driven overexpression or by antisense-mediated downregulation. Clusterin overexpression was associated with an increase in drug-resistance, i.e., with an increased survival of melanoma cells in the presence of cytotoxic drugs. In contrast, downregulation of clusterin by 2'-O-(2-methoxy)ethyl (2'MOE)-modified antisense oligonucleotides (AS-ODN) directed against clusterin mRNA significantly reduced drug-resistance, i.e., decreased survival of melanoma cells in the presence of cytotoxic drugs. To evaluate the effects of clusterin-antisense treatment in vivo, we applied an SCID-mouse/human-melanoma xenotransplantation model. Pre-treatment of mice with the 2'MOE-modified clusterin AS-ODN was associated with a significantly improved tumor response to dacarbazine as compared with animals pretreated with a scrambled control oligonucleotide. Taken together, we show that clusterin is strongly expressed in melanoma. Downregulation of clusterin reduces drug-resistance, i.e., reduces melanoma cell survival in response to cytotoxic drugs in vitro and in vivo. Thus, reducing clusterin expression may provide a novel tool to overcome drug-resistance in melanoma.


Asunto(s)
Resistencia a Antineoplásicos , Glicoproteínas/metabolismo , Melanoma/fisiopatología , Chaperonas Moleculares/metabolismo , Animales , Antineoplásicos/farmacología , Antineoplásicos Alquilantes/farmacología , Apoptosis , Línea Celular Tumoral , Clusterina , Dacarbazina/farmacología , Regulación hacia Abajo , Glicoproteínas/genética , Humanos , Melanocitos/metabolismo , Melanoma/patología , Ratones , Ratones SCID , Chaperonas Moleculares/genética , Trasplante de Neoplasias , Oligonucleótidos Antisentido/farmacología , Trasplante Heterólogo
17.
Oncotarget ; 6(27): 24361-75, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26172298

RESUMEN

All multicellular organisms require a life-long regulation of the number and the size of cells, which build up their organs. mTOR acts as a signaling nodule for the regulation of protein synthesis and growth. To activate the translational cascade, mTOR phosphorylates S6 kinase (S6K1), which is liberated from the eIF3-complex and mobilized for activation of its downstream targets. How S6K1 regulates cell size remains unclear. Here, we challenged cell size control through S6K1 by specifically depleting its binding partner eIF3 in normal and transformed cell lines. We show that loss of eIF3 leads to a massive reduction of cell size and cell number accompanied with an unexpected increase in S6K1-activity. The hyperactive S6K1-signaling was rapamycin-sensitive, suggesting an upstream mTOR-regulation. A selective S6K1 inhibitor (PF-4708671) was unable to interfere with the reduced size, despite efficiently inhibiting S6K1-activity. Restoration of eIF3 expression recovered size defects, without affecting the p-S6 levels. We further show that two, yet uncharacterized, cancer-associated mutations in the eIF3-complex, have the capacity to recover from reduced size phenotype, suggesting a possible role for eIF3 in regulating cancer cell size. Collectively, our results uncover a role for eIF3-complex in maintenance of normal and neoplastic cell size - independent of S6K1-signaling.


Asunto(s)
Tamaño de la Célula , Factor 3 de Iniciación Eucariótica/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Proliferación Celular , Transformación Celular Neoplásica , Inhibidores Enzimáticos/química , Fibroblastos/metabolismo , Células HEK293 , Humanos , Imidazoles/química , Mutación , Fenotipo , Fosforilación , Piperazinas/química , ARN Interferente Pequeño/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/antagonistas & inhibidores , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo
18.
PLoS One ; 7(6): e40078, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22761949

RESUMEN

Cell lines are key tools in cancer research allowing the generation of neoplasias in animal models resembling the initial tumours able to mimic the original neoplasias closely in vivo. Canine lymphoma is the major hematopoietic malignancy in dogs and considered as a valuable spontaneous large animal model for human Non-Hodgkin's Lymphoma (NHL). Herein we describe the establishment and characterisation of an in vivo model using the canine B-cell lymphoma cell line CLBL-1 analysing the stability of the induced tumours and the ability to resemble the original material. CLBL-1 was injected into Rag2(-/-)γ(c) (-/-) mice. The generated tumor material was analysed by immunophenotyping and histopathology and used to establish the cell line CLBL-1M. Both cell lines were karyotyped for detection of chromosomal aberrations. Additionally, CLBL-1 was stimulated with IL-2 and DSP30 as described for primary canine B-cell lymphomas and NHL to examine the stimulatory effect on cell proliferation. CLBL-1 in vivo application resulted in lymphoma-like disease and tumor formation. Immunophenotypic analysis of tumorous material showed expression of CD45(+), MHCII(+), CD11a(+) and CD79αcy(+). PARR analysis showed positivity for IgH indicating a monoclonal character. These cytogenetic, molecular, immunophenotypical and histological characterisations of the in vivo model reveal that the induced tumours and thereof generated cell line resemble closely the original material. After DSP30 and IL-2 stimulation, CLBL-1 showed to respond in the same way as primary material. The herein described CLBL-1 in vivo model provides a highly stable tool for B-cell lymphoma research in veterinary and human medicine allowing various further in vivo studies.


Asunto(s)
Modelos Animales de Enfermedad , Linfoma de Células B/patología , Animales , Línea Celular Tumoral , Perros , Inmunofenotipificación , Ratones , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa
19.
EMBO Mol Med ; 4(4): 283-97, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22323443

RESUMEN

The large difference in phenotypes among tumour populations may stem from the stochastic origin of tumours from distinct cells - tumour cells are assumed to retain the phenotypes of the cells from which they derive. Yet, functional studies addressing the cellular origin of leukaemia are lacking. Here we show that the cells of origin of both, BCR/ABL-induced chronic myeloid (CML) and B-cell acute lymphoid leukaemia (B-ALL), resemble long-term haematopoietic stem cells (LT-HSCs). During disease-maintenance, CML LT-HSCs persist to function as cancer stem cells (CSCs) that maintain leukaemia and require signalling by the transcription factor STAT5. In contrast, B-ALL LT-HSCs differentiate into CSCs that correspond to pro-B cells. This transition step requires a transient IL-7 signal and is lost in IL-7Rα-deficient cells. Thus, in BCR/ABLp185(+) B-ALL and BCR/ABLp210(+) CML, the final phenotype of the tumour as well as the abundance of CSCs is dictated by diverging differentiation fates of their common cells of origin.


Asunto(s)
Transformación Celular Neoplásica/patología , Leucemia Basofílica Aguda/patología , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Células Madre Neoplásicas/patología , Linfocitos B/citología , Linfocitos B/metabolismo , Linfocitos B/patología , Transformación Celular Neoplásica/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Humanos , Leucemia Basofílica Aguda/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Células Madre Neoplásicas/metabolismo , Factor de Transcripción STAT5/metabolismo
20.
EMBO Mol Med ; 2(3): 98-110, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20201032

RESUMEN

Tumourigenesis caused by the Bcr/Abl oncoprotein is a multi-step process proceeding from initial to tumour-maintaining events and finally results in a complex tumour-supporting network. A key to successful cancer therapy is the identification of critical functional nodes in an oncogenic network required for disease maintenance. So far, the transcription factors Stat3 and Stat5a/b have been implicated in bcr/abl-induced initial transformation. However, to qualify as a potential drug target, a signalling pathway must be required for the maintenance of the leukaemic state. Data on the roles of Stat3 or Stat5a/b in leukaemia maintenance are elusive. Here, we show that both, Stat3 and Stat5 are necessary for initial transformation. However, Stat5- but not Stat3-deletion induces G(0)/G(1) cell cycle arrest and apoptosis of imatinib-sensitive and imatinib-resistant stable leukaemic cells in vitro. Accordingly, Stat5-abrogation led to effective elimination of myeloid and lymphoid leukaemia maintenance in vivo. Hence, we identified Stat5 as a vulnerable point in the oncogenic network downstream of Bcr/Abl representing a case of non-oncogene addiction (NOA).


Asunto(s)
Leucemia/fisiopatología , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT5/metabolismo , Animales , Apoptosis , Ciclo Celular , Eliminación de Gen , Genes abl , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Proto-Oncogénicas c-bcr/genética , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT5/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA