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.
J Immunol ; 209(1): 77-92, 2022 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-35705252

RESUMEN

The zinc-finger transcription factor GATA-3 plays a crucial role during early T cell development and also dictates later T cell differentiation outcomes. However, its role and collaboration with the Notch signaling pathway in the induction of T lineage specification and commitment have not been fully elucidated. We show that GATA-3 deficiency in mouse hematopoietic progenitors results in an early block in T cell development despite the presence of Notch signals, with a failure to upregulate Bcl11b expression, leading to a diversion along a myeloid, but not a B cell, lineage fate. GATA-3 deficiency in the presence of Notch signaling results in the apoptosis of early T lineage cells, as seen with inhibition of CDK4/6 (cyclin-dependent kinases 4 and 6) function, and dysregulated cyclin-dependent kinase inhibitor 2b (Cdkn2b) expression. We also show that GATA-3 induces Bcl11b, and together with Bcl11b represses Cdkn2b expression; however, loss of Cdkn2b failed to rescue the developmental block of GATA-3-deficient T cell progenitor. Our findings provide a signaling and transcriptional network by which the T lineage program in response to Notch signals is realized.


Asunto(s)
Factor de Transcripción GATA3/metabolismo , Transducción de Señal , Linfocitos T , Animales , Diferenciación Celular , Linaje de la Célula , Proteínas Inhibidoras de las Quinasas Dependientes de la Ciclina , Redes Reguladoras de Genes , Ratones , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Linfocitos T/metabolismo , Proteínas Supresoras de Tumor/metabolismo
2.
Biochim Biophys Acta Mol Cell Res ; 1864(12): 2449-2459, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28964849

RESUMEN

Although Hematopoietic Stem and Progenitor Cell (HSPC) proliferation, survival and expansion have been shown to be supported by the cooperative action of different cytokines, little is known about the intracellular signaling pathways that are activated by cytokines upon binding to their receptors. Our study showed that Growth factor receptor-bound protein 2 (Grb2) mRNAs are preferentially expressed in HSC compared to progenitors and differentiated cells of the myeloid and erythroid lineages. Conditional deletion of Grb2 induced a rapid decline of erythroid and myeloid progenitors and a progressive decline of HSC numbers in steady state conditions. We showed that when transplanted, Grb2 deleted bone marrow cells could not reconstitute irradiated recipients. Strinkingly, Grb2 deletion did not modify HSPC quiescence, but impaired LT-HSC and progenitors ability to respond a proliferative signal induced by 5FU in vivo and by various cytokines in vitro. We showed finally that Grb2 links IL3 signaling to the ERK/MAPK proliferative pathway and that both SH2 and SH3 domains of Grb2 are crucial for IL3 signaling in progenitor cells. Our findings position Grb2 as a key adaptor that integrates various cytokines response in cycling HSPC.


Asunto(s)
Diferenciación Celular/genética , Linaje de la Célula/genética , Proteína Adaptadora GRB2/genética , Células Madre Hematopoyéticas/metabolismo , Animales , Células de la Médula Ósea/metabolismo , Trasplante de Médula Ósea , Proliferación Celular/genética , Células Eritroides/metabolismo , Técnicas de Inactivación de Genes , Células Madre Hematopoyéticas/citología , Ratones , Células Mieloides/metabolismo , Transducción de Señal
3.
Cell Stem Cell ; 16(3): 302-13, 2015 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-25704240

RESUMEN

Regulated blood production is achieved through the hierarchical organization of dormant hematopoietic stem cell (HSC) subsets that differ in self-renewal potential and division frequency, with long-term (LT)-HSCs dividing the least. The molecular mechanisms underlying this variability in HSC division kinetics are unknown. We report here that quiescence exit kinetics are differentially regulated within human HSC subsets through the expression level of CDK6. LT-HSCs lack CDK6 protein. Short-term (ST)-HSCs are also quiescent but contain high CDK6 protein levels that permit rapid cell cycle entry upon mitogenic stimulation. Enforced CDK6 expression in LT-HSCs shortens quiescence exit and confers competitive advantage without impacting function. Computational modeling suggests that this independent control of quiescence exit kinetics inherently limits LT-HSC divisions and preserves the HSC pool to ensure lifelong hematopoiesis. Thus, differential expression of CDK6 underlies heterogeneity in stem cell quiescence states that functionally regulates this highly regenerative system.


Asunto(s)
División Celular/fisiología , Simulación por Computador , Quinasa 6 Dependiente de la Ciclina/biosíntesis , Regulación Enzimológica de la Expresión Génica/fisiología , Células Madre Hematopoyéticas/enzimología , Modelos Biológicos , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/citología , Humanos
4.
Nat Med ; 20(1): 29-36, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24292392

RESUMEN

Tumor recurrence following treatment remains a major clinical challenge. Evidence from xenograft models and human trials indicates selective enrichment of cancer-initiating cells (CICs) in tumors that survive therapy. Together with recent reports showing that CIC gene signatures influence patient survival, these studies predict that targeting self-renewal, the key 'stemness' property unique to CICs, may represent a new paradigm in cancer therapy. Here we demonstrate that tumor formation and, more specifically, human colorectal CIC function are dependent on the canonical self-renewal regulator BMI-1. Downregulation of BMI-1 inhibits the ability of colorectal CICs to self-renew, resulting in the abrogation of their tumorigenic potential. Treatment of primary colorectal cancer xenografts with a small-molecule BMI-1 inhibitor resulted in colorectal CIC loss with long-term and irreversible impairment of tumor growth. Targeting the BMI-1-related self-renewal machinery provides the basis for a new therapeutic approach in the treatment of colorectal cancer.


Asunto(s)
Neoplasias Colorrectales/tratamiento farmacológico , Compuestos Heterocíclicos con 2 Anillos/farmacología , Recurrencia Local de Neoplasia/metabolismo , Células Madre Neoplásicas/metabolismo , Complejo Represivo Polycomb 1/metabolismo , Tiazoles/farmacología , Animales , Western Blotting , Bromodesoxiuridina , Línea Celular Tumoral , Citometría de Flujo , Vectores Genéticos/genética , Compuestos Heterocíclicos con 2 Anillos/uso terapéutico , Humanos , Luciferasas , Ratones Endogámicos NOD , Ratones SCID , Complejo Represivo Polycomb 1/antagonistas & inhibidores , Interferencia de ARN , ARN Interferente Pequeño/genética , Tiazoles/uso terapéutico
5.
Proc Natl Acad Sci U S A ; 110(51): 20599-604, 2013 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-24297922

RESUMEN

The Lnk (Sh2b3) adaptor protein dampens the response of hematopoietic stem cells and progenitors (HSPCs) to a variety of cytokines by inhibiting JAK2 signaling. As a consequence, Lnk(-/-) mice develop hematopoietic hyperplasia, which progresses to a phenotype resembling the nonacute phase of myeloproliferative neoplasm. In addition, Lnk mutations have been identified in human myeloproliferative neoplasms and acute leukemia. We find that Lnk suppresses the development of radiation-induced acute B-cell malignancies in mice. Lnk-deficient HSPCs recover more effectively from irradiation than their wild-type counterparts, and this resistance of Lnk(-/-) HSPCs to radiation underlies the subsequent emergence of leukemia. A search for the mechanism responsible for radiation resistance identified the cytokine IL-11 as being critical for the ability of Lnk(-/-) HSPCs to recover from irradiation and subsequently become leukemic. In IL-11 signaling, wild-type Lnk suppresses tyrosine phosphorylation of the Src homology region 2 domain-containing phosphatase-2/protein tyrosine phosphatase nonreceptor type 11 and its association with the growth factor receptor-bound protein 2, as well as activation of the Erk MAP kinase pathway. Indeed, Src homology region 2 domain-containing phosphatase-2 has a binding motif for the Lnk Src Homology 2 domain that is phosphorylated in response to IL-11 stimulation. IL-11 therefore drives a pathway that enhances HSPC radioresistance and radiation-induced B-cell malignancies, but is normally attenuated by the inhibitory adaptor Lnk.


Asunto(s)
Rayos gamma/efectos adversos , Interleucina-11/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Leucemia de Células B/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de la radiación , Proteínas de Neoplasias/metabolismo , Neoplasias Inducidas por Radiación/metabolismo , Proteínas/metabolismo , Tolerancia a Radiación/efectos de la radiación , Proteínas Adaptadoras Transductoras de Señales , Secuencias de Aminoácidos , Animales , Proteína Adaptadora GRB2/genética , Proteína Adaptadora GRB2/metabolismo , Humanos , Interleucina-11/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Leucemia de Células B/genética , Leucemia de Células B/patología , Sistema de Señalización de MAP Quinasas/genética , Proteínas de la Membrana , Ratones , Ratones Noqueados , Proteínas de Neoplasias/genética , Neoplasias Inducidas por Radiación/genética , Neoplasias Inducidas por Radiación/patología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Proteínas/genética , Tolerancia a Radiación/genética
6.
Nat Immunol ; 14(10): 1037-44, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23974957

RESUMEN

The transcription factor GATA-3 is expressed and required for differentiation and function throughout the T lymphocyte lineage. Despite evidence it may also be expressed in multipotent hematopoietic stem cells (HSCs), any role for GATA-3 in these cells has remained unclear. Here we found GATA-3 was in the cytoplasm in quiescent long-term stem cells from steady-state bone marrow but relocated to the nucleus when HSCs cycled. Relocation depended on signaling via the mitogen-activated protein kinase p38 and was associated with a diminished capacity for long-term reconstitution after transfer into irradiated mice. Deletion of Gata3 enhanced the repopulating capacity and augmented the self-renewal of long-term HSCs in cell-autonomous fashion without affecting the cell cycle. Our observations position GATA-3 as a regulator of the balance between self-renewal and differentiation in HSCs that acts downstream of the p38 signaling pathway.


Asunto(s)
Factor de Transcripción GATA3/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Animales , Factor de Transcripción GATA3/genética , Eliminación de Gen , Expresión Génica , Hematopoyesis/genética , Células Madre Hematopoyéticas/efectos de los fármacos , Ligandos , Ratones , Ratones Noqueados , Poli I-C/farmacología , Transporte de Proteínas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal , Receptores Toll-Like/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
7.
PLoS One ; 8(3): e58787, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23516556

RESUMEN

Tachykinins are a large group of neuropeptides with both central and peripheral activity. Despite the increasing number of studies reporting a growth supportive effect of tachykinin peptides in various in vitro stem cell systems, it remains unclear whether these findings are applicable in vivo. To determine how neurokinin-1 receptor (NK-1R) deficient hematopoietic stem cells would behave in a normal in vivo environment, we tested their reconstitution efficiency using competitive bone marrow repopulation assays. We show here that bone marrow taken from NK-1R deficient mice (Tacr1(-/-)) showed lineage specific B and T cell engraftment deficits compared to wild-type competitor bone marrow cells, providing evidence for an involvement of NK-1R signalling in adult hematopoiesis. Tachykinin knockout mice lacking the peptides SP and/or HK-1 (Tac1 (-/-), Tac4 (-/-) and Tac1 (-/-)/Tac4 (-/-) mice) repopulated a lethally irradiated wild-type host with similar efficiency as competing wild-type bone marrow. The difference between peptide and receptor deficient mice indicates a paracrine and/or endocrine mechanism of action rather than autocrine signalling, as tachykinin peptides are supplied by the host environment.


Asunto(s)
Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Receptores de Neuroquinina-1/metabolismo , Transducción de Señal , Animales , Linfocitos B/citología , Linfocitos B/metabolismo , Linaje de la Célula , Femenino , Técnicas de Inactivación de Genes , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Ratones , Ratones Endogámicos C57BL , Precursores de Proteínas/deficiencia , Receptores de Neuroquinina-1/deficiencia , Receptores de Neuroquinina-1/genética , Sustancia P/deficiencia , Linfocitos T/citología , Linfocitos T/metabolismo , Taquicininas/deficiencia
8.
Cell Stem Cell ; 6(1): 48-58, 2010 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-20074534

RESUMEN

Sustained blood cell production depends on divisions by hematopoietic stem cells (HSCs) that yield both differentiating progeny as well as new HSCs via self-renewal. Differentiating progeny remain capable of self-renewal, but only HSCs sustain self-renewal through successive divisions securely enough to maintain clones that persist life-long. Until recently, the first identified next stage consisted of "short-term" reconstituting cells able to sustain clones of differentiating cells for only 4-6 weeks. Here we expand evidence for a numerically dominant "intermediate-term" multipotent HSC stage in mice whose clones persist for 6-8 months before becoming extinct and that are separable from both short-term as well as permanently reconstituting "long-term" HSCs. The findings suggest that the first step in stem cell differentiation consists not in loss of initial capacity for serial self-renewal divisions, but rather in loss of mechanisms that stabilize self-renewing behavior throughout successive future stem cell divisions.


Asunto(s)
Diferenciación Celular , División Celular , Células Madre Hematopoyéticas/citología , Animales , Antígenos CD/genética , Antígenos CD34/genética , Linaje de la Célula , Separación Celular , Células Cultivadas , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Células Madre Hematopoyéticas/metabolismo , Integrina alfa2/genética , Ratones , Ratones Endogámicos C57BL , Receptores de Superficie Celular/genética , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria , Factores de Tiempo , Tirosina Quinasa 3 Similar a fms/genética
9.
Int J Cancer ; 125(2): 308-17, 2009 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-19378338

RESUMEN

The Bcr-Abl inhibitor imatinib is the current first-line therapy for all newly diagnosed chronic myeloid leukemia (CML). Nevertheless, resistance to imatinib emerges as CML progresses to an acute deadly phase implying that physiopathologically relevant cellular targets should be validated to develop alternative therapeutic strategies. The NF-kappaB transcription factor that exerts pro-survival actions is found abnormally active in numerous hematologic malignancies. In the present study, using Bcr-Abl-transfected BaF murine cells, LAMA84 human CML cell line and primary CML, we show that NF-kappaB is active downstream of Bcr-Abl. Pharmacological blockade of NF-kappaB by the IKK2 inhibitor AS602868 prevented survival of BaF cells expressing either wild-type, M351T or T315I imatinib-resistant mutant forms of Bcr-Abl both in vitro and in vivo using a mouse xenograft model. AS602868 also affected the survival of LAMA84 cells and of an imatinib-resistant variant. Importantly, the IKK2 inhibitor strongly decreased in vitro survival and ability to form hematopoietic colonies of primary imatinib resistant CML cells including T315I cells. Our data strongly support the targeting of NF-kappaB as a promising new therapeutic opportunity for the treatment of imatinib resistant CML patients in particular in the case of T315I patients. The T315I mutation escapes all currently used Bcr-Abl inhibitors and is likely to become a major clinical problem as it is associated with a poor clinical outcome.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Genes abl/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Mutación , FN-kappa B/antagonistas & inhibidores , Piperazinas/farmacología , Pirimidinas/farmacología , Animales , Benzamidas , Línea Celular Tumoral , Técnica del Anticuerpo Fluorescente , Humanos , Mesilato de Imatinib , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Ratones , Ratones Desnudos
10.
Proc Natl Acad Sci U S A ; 105(51): 20286-90, 2008 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-19095794

RESUMEN

The properties and biology of mRNA transcripts can be affected profoundly by the choice of alternative polyadenylation sites, making definition of the 3' ends of transcripts essential for understanding their regulation. Here we show that 22-52% of sequences in commonly used human and murine "full-length" transcript databases may not currently end at bona fide polyadenylation sites. To identify probable transcript termini over the entire murine and human genomes, we analyzed the EST databases for positional clustering of EST ends. The analysis yielded 58,282 murine- and 86,410 human-candidate polyadenylation sites, of which 75% mapped to 23,091 known murine transcripts and 22,891 known human transcripts. The murine dataset correctly predicted 97% of the 3' ends in a manually curated and experimentally supported benchmark transcript set. Of currently known genes, 15% had no associated prediction and 25% had only a single predicted termination site. The remaining genes had an average of 3-4 alternative polyadenylation sites predicted for each murine or human transcript, respectively. The results are made available in the form of tables and an interactive web site that can be mined for rapid assessment of the validity of 3' ends in existing collections, enumeration of potential alternative 3' polyadenylation sites of known transcripts, direct retrieval of terminal sequences for design of probes, and detection of polyadenylation sites not currently mapped to known genes.


Asunto(s)
Región de Flanqueo 3' , Análisis por Conglomerados , Etiquetas de Secuencia Expresada , Animales , Humanos , Métodos , Ratones , Poli A , Poliadenilación
11.
Cell Signal ; 18(4): 449-55, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15967637

RESUMEN

Survival and proliferation of Hodgkin lymphoma (HL) cells are influenced by many cytokines produced by different cell types in the lymph node microenvironment. STAT, family of transcription factors, are key mediators of cytokine signaling and their perturbation contributes to various human diseases. Electrophoretic mobility shift and phosphoprotein immunoblotting analyses were used to study STAT activation in HL cell lines. We thus observed high levels of constitutively activated STAT1, 3, 5 and 6 in HDLM-2 and L540 cells, which could be correlated with JAK kinase activation. In contrast KM-H2 cells did not display STAT activation. Preventing constitutive STAT activation by specific JAK kinases inhibitors induced apoptosis of HL cell lines and was associated with a strong decrease in the expression of the anti-apoptotic genes IAP-1, IAP-2, Bcl-xL, Bfl1 and Traf1. Silencing of JAKs by specific siRNAs also induced apoptosis of HL cells. Altogether, these results suggest that aberrant STAT activation in Hodgkin cells may promote cell survival and as a consequence facilitate oncogenic transformation.


Asunto(s)
Enfermedad de Hodgkin/metabolismo , Factores de Transcripción STAT/metabolismo , Apoptosis/efectos de los fármacos , Western Blotting , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Ensayo de Cambio de Movilidad Electroforética , Humanos , Inmunoprecipitación , Quinazolinas/farmacología , ARN Interferente Pequeño/farmacología , Factores de Transcripción STAT/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Células Tumorales Cultivadas , Tirfostinos/farmacología
12.
Blood ; 105(2): 804-11, 2005 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-15454494

RESUMEN

Acute myeloid leukemia (AML) cells are characterized by a constitutive and abnormal activation of the nuclear factor-kappaB (NF-kappaB) transcription factor. This study, conducted in vitro on 18 patients, shows that targeting the IKB kinase 2 (IKK2) kinase with the specific pharmacologic inhibitor AS602868 to block NF-kappaB activation led to apoptosis of human primary AML cells. Moreover, AS602868 potentiated the apoptotic response induced by the current chemotherapeutic drugs doxorubicin, cytarabine, or etoposide (VP16). AS602868-induced cell death was associated with rupture of the mitochondrial transmembrane potential and activation of cellular caspases. NF-kappaB inhibition did not affect normal CD34+ hematopoietic precursors, suggesting that it could represent a new adjuvant strategy for AML treatment.


Asunto(s)
Apoptosis/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Leucemia Mieloide , FN-kappa B/metabolismo , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Enfermedad Aguda , Adulto , Anciano , Anciano de 80 o más Años , Antígenos CD34/metabolismo , Antineoplásicos Fitogénicos/farmacología , Caspasas/metabolismo , Niño , Resistencia a Antineoplásicos , Etopósido/farmacología , Femenino , Humanos , Quinasa I-kappa B , Masculino , Persona de Mediana Edad , FN-kappa B/antagonistas & inhibidores , Fosforilación/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/enzimología , Células Tumorales Cultivadas/metabolismo
13.
Oncogene ; 22(50): 8187-94, 2003 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-14603259

RESUMEN

NF-kappaB transcription factors promote survival in numerous cell types via induction of antiapoptotic genes. Pharmacological blockade of the IKK2 kinase with AS602868, a specific inhibitor that competes with ATP binding, prevented TNF-alpha-induced NF-kappaB activation in Jurkat leukemic T cells. While TNF-alpha by itself had no effect on Jurkat survival, the addition of AS602868 induced cell death, visualized by DNA fragmentation and sub-G1 analysis. A disruption of the mitochondrial potential followed by activation of caspases 9 and 3 was observed in cells treated by the combination TNF-alpha+AS602868. Quantitative real-time PCR demonstrated that AS602868 prevented TNF-alpha induction of the antiapoptotic genes coding for c-IAP-2, Bclx, Bfl-1/A1 and Traf-1. The use of a specific IKK2 inhibitor appears, therefore, as an interesting pharmaceutical strategy to increase the cell's sensitivity towards apoptotic effectors.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Leucemia de Células T/tratamiento farmacológico , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/farmacología , Caspasa 3 , Caspasa 9 , Caspasas/metabolismo , Caspasas/fisiología , Humanos , Quinasa I-kappa B , Células Jurkat , Potenciales de la Membrana/fisiología , Mitocondrias/fisiología , Acetato de Tetradecanoilforbol/metabolismo
14.
Mol Diagn ; 7(3-4): 187-94, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-15068390

RESUMEN

BACKGROUND: Nuclear factor-kappa B (NF-kappa B) is an important transcription factor involved in the regulation of immune responses as well as in cell proliferation and survival. An abnormal and constitutive activation of NF-kappa B is observed in many pathological states as diverse as inflammation, neurological diseases, and cancer. METHODS AND RESULTS: Termination of NF-kappa B transcription is mediated through the NF-kappa B-dependent synthesis of the I kappa B-alpha inhibitory subunit. To quantify NF-kappa B activation we measured by real-time PCR the expression of I kappa B-alpha mRNA. The PCR data perfectly matched the results obtained by Northern blot or gene reporter analysis when Jurkat leukemic T cells or HeLa carcinoma cells were stimulated with various activators of NF-kappa B, such as the cytokine tumor necrosis factor (TNF)-alpha or the phorbol ester PMA. Constitutive NF-kappa B activation in Hodgkin's lymphoma cell line could also be evaluated by this approach. Kinetic experiments in HeLa cells show that TNF stimulation first induced NF-kappa B DNA binding within 30 minutes, followed by I kappa B-alpha gene transcription 30 minutes later. Removal of TNF after stimulation resulted in a faster decrease in both NF-kappa B DNA binding activity and I kappa B-alpha mRNA levels. No accumulation or stabilization of I kappa B-alpha mRNA was detected that could bias interpretation of the results. The sensitivity of the method allowed the detection of NF-kappa B activation in stimulated normal peripheral blood lymphocytes. CONCLUSION: The real-time PCR measure of I kappa B-alpha mRNA levels is a rapid, sensitive, and powerful method to quantify the transcriptional power of NF-kappa B. It can be easily used for clinical evaluation of NF-kappa B status.


Asunto(s)
Proteínas I-kappa B/genética , FN-kappa B/genética , Reacción en Cadena de la Polimerasa/métodos , Activación Transcripcional/genética , Secuencia de Bases , Cartilla de ADN , Regulación Neoplásica de la Expresión Génica/genética , Células HeLa , Humanos , Células Jurkat , Inhibidor NF-kappaB alfa , FN-kappa B/metabolismo , ARN Mensajero/genética , Transcripción Genética
15.
Oncogene ; 21(20): 3213-24, 2002 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-12082637

RESUMEN

The transcription factor NF-kappaB promotes cell survival. Using a variant of Jurkat leukemic T cells expressing IkappaB-alphaDeltaN, a super-repressor of NF-kappaB activation we first show that the tumor promoter PMA could prevent Fas-induced apoptosis via activation of NF-kappaB. Moreover, we demonstrate that in the absence of NF-kappaB activation, PMA became a strong inducer of apoptosis through stimulation of the upstream caspases 8 and 9 as well as of the effector caspase 3. A RNase-protection analysis showed that PMA stimulated the expression of several known anti-apoptotic genes (TRAF1, TRAF4, c-IAP-1, c-IAP-2, Bfl-1, Bcl-xl). In the absence of NF-kappaB activation, these survival influences were strongly lowered revealing the apoptotic effect of PMA. These results suggest that NF-kappaB activation could be an important step in the tumor promoting effect of PMA.


Asunto(s)
Apoptosis/efectos de los fármacos , Proteínas de Unión al ADN/fisiología , Regulación Leucémica de la Expresión Génica/fisiología , Proteínas I-kappa B , FN-kappa B/fisiología , Proteínas de Neoplasias/biosíntesis , Acetato de Tetradecanoilforbol/farmacología , Apoptosis/genética , Carbazoles/farmacología , Caspasa 8 , Caspasa 9 , Caspasas/metabolismo , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Proteína Ligando Fas , Humanos , Indoles/farmacología , Isoenzimas/antagonistas & inhibidores , Isoenzimas/fisiología , Células Jurkat/efectos de los fármacos , Células Jurkat/metabolismo , Maleimidas/farmacología , Glicoproteínas de Membrana/fisiología , Inhibidor NF-kappaB alfa , Proteínas de Neoplasias/genética , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/fisiología , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/fisiología , Eliminación de Secuencia , Receptor fas/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...