Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Immunol ; 25(6): 994-1006, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38671323

RESUMEN

The lung is constantly exposed to the outside world and optimal adaptation of immune responses is crucial for efficient pathogen clearance. However, mechanisms that lead to lung-associated macrophages' functional and developmental adaptation remain elusive. To reveal such mechanisms, we developed a reductionist model of environmental intranasal ß-glucan exposure, allowing for the detailed interrogation of molecular mechanisms of pulmonary macrophage adaptation. Employing single-cell transcriptomics, high-dimensional imaging and flow cytometric characterization paired with in vivo and ex vivo challenge models, we reveal that pulmonary low-grade inflammation results in the development of apolipoprotein E (ApoE)-dependent monocyte-derived alveolar macrophages (ApoE+CD11b+ AMs). ApoE+CD11b+ AMs expressed high levels of CD11b, ApoE, Gpnmb and Ccl6, were glycolytic, highly phagocytic and produced large amounts of interleukin-6 upon restimulation. Functional differences were cell intrinsic, and myeloid cell-specific ApoE ablation inhibited Ly6c+ monocyte to ApoE+CD11b+ AM differentiation dependent on macrophage colony-stimulating factor secretion, promoting ApoE+CD11b+ AM cell death and thus impeding ApoE+CD11b+ AM maintenance. In vivo, ß-glucan-elicited ApoE+CD11b+ AMs limited the bacterial burden of Legionella pneumophilia after infection and improved the disease outcome in vivo and ex vivo in a murine lung fibrosis model. Collectively these data identify ApoE+CD11b+ AMs generated upon environmental cues, under the control of ApoE signaling, as an essential determinant for lung adaptation enhancing tissue resilience.


Asunto(s)
Apolipoproteínas E , Lectinas Tipo C , Macrófagos Alveolares , Ratones Endogámicos C57BL , beta-Glucanos , Animales , Ratones , Adaptación Fisiológica/inmunología , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Antígeno CD11b/metabolismo , Diferenciación Celular , Lectinas Tipo C/metabolismo , Pulmón/inmunología , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/metabolismo , Ratones Noqueados , Monocitos/inmunología , Monocitos/metabolismo
2.
Oncogenesis ; 5(12): e278, 2016 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-27941931

RESUMEN

Hypoxia-inducible factor 1α (Hif1α) is a key regulator of cellular adaptation and survival under hypoxic conditions. In pancreatic ductal adenocarcinoma (PDAC), it has been recently shown that genetic ablation of Hif1α accelerates tumour development by promoting tumour-supportive inflammation in mice, questioning its role as the key downstream target of many oncogenic signals of PDAC. Likely, Hif1α has a context-dependent role in pancreatic tumorigenesis. To further analyse this, murine PDAC cell lines with reduced Hif1α expression were generated using shRNA transfection. Cells were transplanted into wild-type mice through orthotopic or portal vein injection in order to test the in vivo function of Hif1α in two major tumour-associated biological scenarios: primary tumour growth and remote colonization/metastasis. Although Hif1α protects PDAC cells from stress-induced cell deaths in both scenarios-in line with the general function Hif1α-its depletion leads to different oncogenic consequences. Hif1α depletion results in rapid tumour growth with marked hypoxia-induced cell death, which potentially leads to a persistent tumour-sustaining inflammatory response. However, it simultaneously reduces tumour colonization and hepatic metastases by increasing the susceptibility to anoikis induced by anchorage-independent conditions. Taken together, the role of Hif1α in pancreatic tumorigenesis is context-dependent. Clinical trials of Hif1α inhibitors need to take this into account, targeting the appropriate scenario, for example palliative vs adjuvant therapy.

3.
Leukemia ; 30(7): 1520-30, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27055871

RESUMEN

T lymphocyte non-Hodgkin's lymphoma (T-NHL) represents an aggressive and largely therapy-resistant subtype of lymphoid malignancies. As deregulated apoptosis is a frequent hallmark of lymphomagenesis, we analyzed gene expression profiles and protein levels of primary human T-NHL samples for various apoptotic regulators. We identified the apoptotic regulator MCL-1 as the only pro-survival BCL-2 family member to be highly expressed throughout all human T-NHL subtypes. Functional validation of pro-survival protein members of the BCL-2 family in two independent T-NHL mouse models identified that the partial loss of Mcl-1 significantly delayed T-NHL development in vivo. Moreover, the inducible reduction of MCL-1 protein levels in lymphoma-burdened mice severely impaired the continued survival of T-NHL cells, increased their susceptibility to chemotherapeutics and delayed lymphoma progression. Lymphoma viability remained unaffected by the genetic deletion or pharmacological inhibition of all alternative BCL-2 family members. Consistent with a therapeutic window for MCL-1 treatment within the context of the whole organism, we observed an only minimal toxicity after systemic heterozygous loss of Mcl-1 in vivo. We conclude that re-activation of mitochondrial apoptosis by blockade of MCL-1 represents a promising therapeutic strategy to treat T-cell lymphoma.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Apoptosis , Linfoma de Células T/química , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/análisis , Animales , Proteínas Reguladoras de la Apoptosis/análisis , Supervivencia Celular , Resistencia a Antineoplásicos , Perfilación de la Expresión Génica , Humanos , Linfoma de Células T/patología , Ratones , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/antagonistas & inhibidores , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/genética , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/fisiología , Proteínas Proto-Oncogénicas c-bcl-2/genética
4.
Oncogene ; 35(32): 4269-81, 2016 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-26776161

RESUMEN

Constitutive activation of the antiapoptotic nuclear factor-κB (NF-κB) signaling pathway is a hallmark of the activated B-cell-like (ABC) subtype of diffuse large B-cell lymphomas (DLBCL). Recurrent oncogenic mutations are found in the scaffold protein CARMA1 (CARD11) that connects B-cell receptor (BCR) signaling to the canonical NF-κB pathway. We asked how far additional downstream processes are activated and contribute to the oncogenic potential of DLBCL-derived CARMA1 mutants. To this end, we expressed oncogenic CARMA1 in the NF-κB negative DLBCL lymphoma cell line BJAB. By a proteomic approach we identified recruitment of ß-catenin and its destruction complex consisting of APC, AXIN1, CK1α and GSK3ß to oncogenic CARMA1. Recruitment of the ß-catenin destruction complex was independent of CARMA1-BCL10-MALT1 complex formation or constitutive NF-κB activation and promoted the stabilization of ß-catenin. The ß-catenin destruction complex was also recruited to CARMA1 in ABC DLBCL cell lines, which coincided with elevated ß-catenin expression. In line, ß-catenin was frequently detected in non-GCB DLBCL biopsies that rely on chronic BCR signaling. Increased ß-catenin amounts alone were not sufficient to induce classical WNT target gene signatures, but could augment TCF/LEF-dependent transcriptional activation in response to WNT signaling. In conjunction with NF-κB, ß-catenin enhanced expression of immunosuppressive interleukin-10 and suppressed antitumoral CCL3, indicating that ß-catenin can induce a favorable tumor microenvironment. Thus, parallel activation of NF-κB and ß-catenin signaling by gain-of-function mutations in CARMA1 augments WNT stimulation and is required for regulating the expression of distinct NF-κB target genes to trigger cell-intrinsic and extrinsic processes that promote DLBCL lymphomagenesis.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD/genética , Proteínas Adaptadoras de Señalización CARD/metabolismo , Carcinogénesis , Guanilato Ciclasa/genética , Guanilato Ciclasa/metabolismo , Linfoma de Células B Grandes Difuso/metabolismo , FN-kappa B/metabolismo , Transducción de Señal , beta Catenina/metabolismo , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Linfoma de Células B Grandes Difuso/patología , Mutación , Estabilidad Proteica , Factores de Transcripción TCF/metabolismo , Proteínas de Unión al GTP rho/metabolismo
5.
Leukemia ; 30(1): 112-23, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26153654

RESUMEN

Deregulated apoptosis is an identifying feature of myelodysplastic syndromes (MDS). Whereas apoptosis is increased in the bone marrow (BM) of low-risk MDS patients, progression to high-risk MDS correlates with an acquired resistance to apoptosis and an aberrant expression of BCL-2 proteins. To overcome the acquired apoptotic resistance in high-risk MDS, we investigated the induction of apoptosis by inhibition of pro-survival BCL-2 proteins using the BCL-2/-XL/-W inhibitor ABT-737 or the BCL-2-selective inhibitor ABT-199. We characterized a cohort of 124 primary human BM samples from MDS/secondary acute myeloid leukemia (sAML) patients and 57 healthy, age-matched controls. Inhibition of anti-apoptotic BCL-2 proteins was specifically toxic for BM cells from high-risk MDS and sAML patients, whereas low-risk MDS or healthy controls remained unaffected. Notably, ABT-737 or ABT-199 treatment was capable of targeting the MDS stem/progenitor compartment in high-risk MDS/sAML samples as shown by the reduction in CD34(+) cells and the decreased colony-forming capacity. Elevated expression of MCL-1 conveyed resistance against both compounds. Protection by stromal cells only partially inhibited induction of apoptosis. Collectively, our data show that the apoptotic resistance observed in high-risk MDS/sAML cells can be overcome by the ABT-737 or ABT-199 treatment and implies that BH3 mimetics might delay disease progression in higher-risk MDS or sAML patients.


Asunto(s)
Apoptosis/efectos de los fármacos , Compuestos de Bifenilo/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Síndromes Mielodisplásicos/tratamiento farmacológico , Nitrofenoles/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Células Madre/efectos de los fármacos , Sulfonamidas/farmacología , Células Cultivadas , Humanos , Síndromes Mielodisplásicos/patología , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/análisis , Piperazinas/farmacología
6.
Curr Drug Targets ; 7(10): 1335-40, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17073595

RESUMEN

The development of lymphomas and leukemias is frequently caused by chromosomal translocations that deregulate cellular pathways of differentiation, proliferation or survival. The molecules that are involved in these aberrations provide rational targets for selective drug therapies. Recently, several disease specific translocations have been identified in human MALT lymphoma. These aberrations either upregulate the expression of BCL10 or MALT1 or induce the formation of API2-MALT1 fusion proteins. Genetic and biochemical experiments identified BCL10 and MALT1 as central components of an oligomerization-ubiquitinylation-phosphorylation cascade that activates the transcription factor NF-kappaB in response to antigen receptor ligation. Deregulation of the signaling cascade is directly associated with antigen independent MALT lymphoma growth. Here we provide an overview of the physiological and pathological functions of BCL10/MALT1 signal transduction and discuss the potential of this pathway as a drug target.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Caspasas/fisiología , Sistemas de Liberación de Medicamentos/métodos , Linfoma/tratamiento farmacológico , Linfoma/metabolismo , Proteínas de Neoplasias/fisiología , Transducción de Señal/efectos de los fármacos , Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Animales , Antineoplásicos/uso terapéutico , Proteína 10 de la LLC-Linfoma de Células B , Inhibidores de Caspasas , Humanos , Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas , Proteínas de Neoplasias/antagonistas & inhibidores , Transducción de Señal/fisiología
7.
J Biol Chem ; 276(33): 30589-97, 2001 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-11387339

RESUMEN

Bcl10 and MALT1, products of distinct chromosomal translocations in mucosa-associated lymphoid tissue lymphoma, cooperate in activating NF-kappaB. Mice lacking Bcl10 demonstrate severe immunodeficiency associated with failure of lymphocytes to activate nuclear factor kappaB (NF-kappaB) in response to antigen receptor stimulation and protein kinase C activation. We characterize Bimp1, a new signaling protein that binds Bcl10 and activates NF-kappaB. Bimp1-mediated NF-kappaB activation requires Bcl10 and IkappaB kinases, indicating that Bimp1 acts upstream of these mediators. Bimp1, Bcl10, and MALT1 form a ternary complex, with Bcl10 bridging the Bimp1/MALT1 interaction. A dominant negative Bimp1 mutant inhibits NF-kappaB activation by anti-CD3 ligation, phorbol ester, and protein kinase C expression. These results suggest that Bimp1 links surface receptor stimulation and protein kinase C activation to Bcl10/MALT1, thus leading to NF-kappaB induction.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Linfoma de Células B de la Zona Marginal , FN-kappa B/biosíntesis , Proteínas de Neoplasias/fisiología , Nucleósido-Fosfato Quinasa/fisiología , Proteína Quinasa C/fisiología , Secuencia de Aminoácidos , Proteína 10 de la LLC-Linfoma de Células B , Caspasas , ADN Complementario/análisis , Activación Enzimática , Guanilato-Quinasas , Humanos , Quinasa I-kappa B , Datos de Secuencia Molecular , Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas , Proteínas de Neoplasias/metabolismo , Proteínas Serina-Treonina Quinasas/fisiología
8.
J Biol Chem ; 276(32): 30461-6, 2001 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-11399756

RESUMEN

Akt is a serine-threonine kinase known to exert antiapoptotic effects through several downstream targets. Akt is cleaved during mitochondrial-mediated apoptosis in a caspase-dependent manner. The reason for this is not clear, however, because Akt has not been demonstrated to be activated in response to mitochondrial apoptotic stimuli. Accordingly, we explored whether the well described mitochondrial apoptotic stimuli staurosporine (STS) and etoposide activate Akt and whether such activation impacts apoptosis. Both STS and etoposide activated Akt in NIH 3T3 cells, maximally at 8 and 2 h, respectively, preceding the onset of apoptosis and poly(ADP-ribose) polymerase cleavage. The overexpression of Akt delayed STS-induced apoptosis with an even more pronounced delay observed with overexpression of constitutively active Akt. Akt activation by proapoptotic stimuli lay upstream of mitochondria, because neither caspase inhibitors nor overexpression of Bcl-2 or Bcl-x(L) could prevent it. Activation depended on phosphatidylinositol 3-kinase activity, however. Conversely, inhibition of phosphatidylinositol 3-kinase with wortmannin sensitized cells to apoptosis initiated by STS. These data demonstrate that mitochondrial apoptotic stimuli also activate Akt and such activation modulates apoptosis in this setting.


Asunto(s)
Apoptosis , Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas/fisiología , Células 3T3 , Androstadienos/farmacología , Animales , Relación Dosis-Respuesta a Droga , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Etopósido/farmacología , Immunoblotting , Ratones , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Unión Proteica , Proteínas Proto-Oncogénicas c-akt , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Estaurosporina/farmacología , Factores de Tiempo , Transfección , Wortmanina , Proteína bcl-X
9.
Cell ; 104(1): 33-42, 2001 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-11163238

RESUMEN

Bcl10, a CARD-containing protein identified from the t(1;14)(p22;q32) breakpoint in MALT lymphomas, has been shown to induce apoptosis and activate NF-kappaB in vitro. We show that one-third of bcl10-/- embryos developed exencephaly, leading to embryonic lethality. Surprisingly, bcl10-/- cells retained susceptibility to various apoptotic stimuli in vivo and in vitro. However, surviving bcl10-/- mice were severely immunodeficient and bcl10-/- lymphocytes are defective in antigen receptor or PMA/Ionomycin-induced activation. Early tyrosine phosphorylation, MAPK and AP-1 activation, and Ca2+ signaling were normal in mutant lymphocytes, but antigen receptor-induced NF-kappaB activation was absent. Thus, Bcl10 functions as a positive regulator of lymphocyte proliferation that specifically connects antigen receptor signaling in B and T cells to NF-kappaB activation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Sistema Nervioso Central/anomalías , FN-kappa B/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/inmunología , Defectos del Tubo Neural/fisiopatología , Receptores de Antígenos/metabolismo , Animales , Anisomicina/farmacología , Formación de Anticuerpos/inmunología , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/inmunología , Apoptosis/efectos de la radiación , Proteína 10 de la LLC-Linfoma de Células B , Linfocitos B/inmunología , División Celular/inmunología , Sistema Nervioso Central/fisiología , Cisplatino/farmacología , Inhibidores Enzimáticos/farmacología , Etopósido/farmacología , Genes Letales/inmunología , Inmunidad Celular/inmunología , Activación de Linfocitos/fisiología , Ratones , Ratones Noqueados , FN-kappa B/inmunología , Proteínas de Neoplasias/metabolismo , Defectos del Tubo Neural/inmunología , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Inhibidores de la Síntesis de la Proteína/farmacología , Receptores de Antígenos/inmunología , Transducción de Señal/inmunología , Estaurosporina/farmacología , Linfocitos T/inmunología
10.
Proc Natl Acad Sci U S A ; 98(4): 1619-24, 2001 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-11172000

RESUMEN

The p53 tumor suppressor protein and the MDM2 oncoprotein form a feedback-control loop that up-regulates cellular MDM2 production, blocks p53 activity, and promotes p53 decay. tsg101 was discovered as a gene whose deficiency results in neoplastic transformation of NIH 3T3 cells and the ability to generate metastatic tumors in nude mice. Its protein product contains a domain, Ubc, characteristic of the catalytic domain of ubiquitin conjugase (E2) enzymes but lacking an active-site cysteine crucial for ubiquitin conjugase activity. Here we report that TSG101 participates with MDM2 in an autoregulatory loop that modulates the cellular levels of both proteins, and also of p53, by affecting protein decay. We show that the Ubc domain of TSG101 interferes with ubiquitination of MDM2, that TSG101 inhibits MDM2 decay and elevates its steady-state level, and that these events are associated with down-regulation of p53 protein. Conversely, pulse-chase and Western blot experiments in wild-type and mutant fibroblasts indicate that elevation of MDM2 by overexpression of wild-type p53, by amplification of the endogenous MDM2 gene, or by transfection of MDM2-expressing constructs promotes TSG101 loss, which we show occurs by 26S proteasome-dependent decay. Our results identify TSG101 as both a regulator of, and target of, MDM2/p53 circuitry.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas Nucleares , Proteínas Proto-Oncogénicas/metabolismo , Factores de Transcripción/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Células 3T3 , Animales , Proteínas de Unión al ADN/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte , Humanos , Ligasas/metabolismo , Ratones , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-mdm2 , Factores de Transcripción/genética , Células Tumorales Cultivadas , Enzimas Ubiquitina-Conjugadoras , Ubiquitinas/metabolismo
11.
Proc Natl Acad Sci U S A ; 98(4): 1859-64, 2001 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-11172041

RESUMEN

Functional inactivation of the tumor susceptibility gene tsg101 in NIH 3T3 fibroblasts results in cellular transformation and the ability to form metastatic tumors in nude mice. The N-terminal region of tsg101 protein is structurally similar to the catalytic domain of ubiquitin-conjugating enzymes, suggesting a potential role of tsg101 in ubiquitin-mediated protein degradation. The C-terminal domain of TSG101 can function as a repressor of transcription. To investigate the physiological function of tsg101, we generated a null mutation of the mouse gene by gene targeting. Homozygous tsg101-/- embryos fail to develop past day 6.5 of embryogenesis (E6.5), are reduced in size, and do not form mesoderm. Mutant embryos show a decrease in cellular proliferation in vivo and in vitro but no increase in apoptosis. Although levels of p53 transcripts were not affected in tsg101-/- embryos, p53 protein accumulated dramatically, implying altered posttranscriptional control of p53. In addition, transcription of the p53 effector, cyclin-dependent kinase inhibitor p21(WAF-1/CIP-1), was increased 5- to 10-fold, whereas activation of MDM2 transcription secondary to p53 elevation was not observed. Introduction of a p53 null mutation into tsg101-/- embryos rescued the gastrulation defect and prolonged survival until E8.5. These results demonstrate that tsg101 is essential for the proliferative burst before the onset of gastrulation and establish a functional connection between tsg101 and the p53 pathway in vivo.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Pérdida del Embrión/metabolismo , Proteínas Nucleares , Factores de Transcripción/fisiología , Proteína p53 Supresora de Tumor/metabolismo , Animales , División Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/metabolismo , Proteínas de Unión al ADN/genética , Desarrollo Embrionario y Fetal , Endodermo/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte , Expresión Génica , Marcación de Gen , Mesodermo/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-mdm2 , Factores de Transcripción/genética , Proteína p53 Supresora de Tumor/genética
12.
EMBO J ; 19(19): 5092-104, 2000 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-11013212

RESUMEN

The hexosamine pathway provides UDP-N:-acetylhexosamine donor substrates used in cytosolic and Golgi-mediated glycosylation of proteins and for formation of glycosylphosphatidylinositol (GPI) anchors, which tether proteins to the outer plasma membrane. We have recently identified the murine glucosamine-6-phosphate (GlcN6P) acetyltransferase, EMeg32, as a developmentally regulated enzyme on the route to UDP-N:-acetylglucosamine (UDP-GlcNAc). Here we describe embryos and cells that have the EMeg32 gene inactivated by homologous recombination. Homozygous mutant embryos die at around embryonic day (E) 7.5 with a general proliferative delay of development. In vitro differentiated EMeg32(-/-) ES cells show reduced proliferation. Mouse embryonic fibroblasts (MEFs) deficient for EMeg32 exhibit defects in proliferation and adhesiveness, which could be complemented by stable re-expression of EMeg32 or by nutritional restoration of intracellular UDP-GlcNAc levels. Reduced UDP-GlcNAc levels predominantly translated into decreased O-GlcNAc modifications of cytosolic and nuclear proteins. Interestingly, growth-impaired EMeg32(-/-) MEFs withstand a number of apoptotic stimuli and express activated PKB/AKT. Thus, EMeg32-dependent UDP-GlcNAc levels influence cell cycle progression and susceptibility to apoptotic stimuli.


Asunto(s)
Acetiltransferasas/metabolismo , Desarrollo Embrionario y Fetal/fisiología , Proteínas de la Membrana/metabolismo , Uridina Difosfato N-Acetilglucosamina/metabolismo , Acetiltransferasas/genética , Animales , Apoptosis , División Celular , Línea Celular , Cromatografía Líquida de Alta Presión , Electroforesis en Gel de Poliacrilamida , Pérdida del Embrión/enzimología , Pérdida del Embrión/metabolismo , Embrión de Mamíferos , Desarrollo Embrionario y Fetal/genética , Retículo Endoplásmico/metabolismo , Fibroblastos/citología , Fibroblastos/enzimología , Fibroblastos/metabolismo , Glucosamina 6-Fosfato N-Acetiltransferasa , Glicosilación , Glicosilfosfatidilinositoles/biosíntesis , Aparato de Golgi/enzimología , Aparato de Golgi/metabolismo , Hibridación in Situ , Etiquetado Corte-Fin in Situ , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa
13.
Science ; 287(5459): 1824-7, 2000 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-10710310

RESUMEN

Chk2 is a protein kinase that is activated in response to DNA damage and may regulate cell cycle arrest. We generated Chk2-deficient mouse cells by gene targeting. Chk2-/- embryonic stem cells failed to maintain gamma-irradiation-induced arrest in the G2 phase of the cell cycle. Chk2-/- thymocytes were resistant to DNA damage-induced apoptosis. Chk2-/- cells were defective for p53 stabilization and for induction of p53-dependent transcripts such as p21 in response to gamma irradiation. Reintroduction of the Chk2 gene restored p53-dependent transcription in response to gamma irradiation. Chk2 directly phosphorylated p53 on serine 20, which is known to interfere with Mdm2 binding. This provides a mechanism for increased stability of p53 by prevention of ubiquitination in response to DNA damage.


Asunto(s)
Daño del ADN , Interfase , Proteínas Nucleares , Proteínas Quinasas , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Apoptosis , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular , Quinasa de Punto de Control 2 , Proteínas de Unión al ADN , Fase G1 , Fase G2 , Rayos gamma , Regulación de la Expresión Génica , Marcación de Gen , Genes Supresores de Tumor , Genes p53 , Humanos , Ratones , Fosforilación , Fosfoserina/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-mdm2 , Células Madre/citología , Células Madre/metabolismo , Linfocitos T/citología , Transcripción Genética , Proteínas Supresoras de Tumor
14.
Cell ; 95(1): 29-39, 1998 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-9778245

RESUMEN

PTEN is a tumor suppressor with sequence homology to protein tyrosine phosphatases and the cytoskeletal protein tensin. mPTEN-mutant mouse embryos display regions of increased proliferation. In contrast, mPTEN-deficient immortalized mouse embryonic fibroblasts exhibit decreased sensitivity to cell death in response to a number of apoptotic stimuli, accompanied by constitutively elevated activity and phosphorylation of protein kinase B/Akt, a crucial regulator of cell survival. Expression of exogenous PTEN in mutant cells restores both their sensitivity to agonist-induced apoptosis and normal pattern of PKB/Akt phosphorylation. Furthermore, PTEN negatively regulates intracellular levels of phosphatidylinositol (3,4,5) trisphosphate in cells and dephosphorylates it in vitro. Our results show that PTEN may exert its role as a tumor suppressor by negatively regulating the PI3'K/PKB/Akt signaling pathway.


Asunto(s)
Apoptosis , Genes Supresores de Tumor , Monoéster Fosfórico Hidrolasas , Proteínas Serina-Treonina Quinasas , Proteínas Tirosina Fosfatasas/fisiología , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Supresoras de Tumor , Animales , División Celular , Supervivencia Celular , Células Cultivadas , Femenino , Fibroblastos/citología , Ratones , Mutagénesis , Fosfohidrolasa PTEN , Fosfatos de Fosfatidilinositol/metabolismo , Proteínas Tirosina Fosfatasas/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-akt , Especificidad por Sustrato
15.
Proc Natl Acad Sci U S A ; 90(8): 3403-7, 1993 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-7682700

RESUMEN

The membrane currents of chicken embryo fibroblasts (CEFs) transformed by Rous sarcoma virus (RSV) were compared with the currents of their nontransformed counterparts by using the whole-cell patch-clamp technique. In nontransformed CEFs, the main membrane current is a delayed outward K+ current that is sensitive to tetraethylammonium ion but insensitive to 4-aminopyridine. This K+ current is almost independent of the intracellular Ca2+ concentration and becomes completely inactivated at positive membrane potentials with a time constant of about 10 s at +30 mV. In contrast, transformed CEFs exhibit a noninactivating K+ current that strongly depends on the intracellular Ca2+ concentration. This Ca(2+)-dependent K+ current is blocked by the scorpion toxin charybdotoxin with an IC50 value of 19 nM, whereas the K+ current of normal CEFs is insensitive to charybdotoxin (up to 300 nM). The K+ current properties of transformed CEFs were also found after microinjection of purified, enzymatically active pp60v-src into normal CEFs but not after infection of CEFs with the Rous-associated virus RAV5, which lacks the v-src oncogene. Our results suggest that the oncogene product pp60v-src modulates existing K+ channel proteins, leading to profound electrophysiological and pharmacological alterations of the K+ current properties in RSV-transformed CEFs. Furthermore, our experiments identify for the first time K+ channels as possible substrates of pp60v-src.


Asunto(s)
Virus del Sarcoma Aviar/genética , Transformación Celular Neoplásica , Proteína Oncogénica pp60(v-src)/metabolismo , Canales de Potasio/fisiología , Animales , Apamina/farmacología , Células Cultivadas , Caribdotoxina , Embrión de Pollo , Venenos Elapídicos/farmacología , Fibroblastos/fisiología , Cinética , Potenciales de la Membrana/efectos de los fármacos , Microinyecciones , Proteína Oncogénica pp60(v-src)/administración & dosificación , Canales de Potasio/efectos de los fármacos , Venenos de Escorpión/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...