Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Cell Death Differ ; 20(2): 321-32, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23059785

RESUMEN

Autophagy is a lysosomal degradation pathway important for cellular homeostasis, mammalian development, cancer and immunity. Many molecular components of autophagy have been identified, but little is known about regulatory mechanisms controlling their effector functions. Here, we show that, in contrast to other p38 MAP kinase activators, the growth arrest and DNA damage 45 beta (Gadd45ß)-MAPK/ERK kinase kinase 4 (MEKK4) pathway specifically directs p38 to autophagosomes. This process results in an accumulation of autophagosomes through p38-mediated inhibition of lysosome fusion. Conversely, autophagic flux is increased in p38-deficient fibroblasts and Gadd45ß-deficient cells. We further identified the underlying mechanism and demonstrate that phosphorylation of the autophagy regulator autophagy-related (Atg)5 at threonine 75 through p38 is responsible for inhibition of starvation-induced autophagy. Thus, we show for the first time that Atg5 activity is controlled by phosphorylation and, moreover, that the spatial regulation of p38 by Gadd45ß/MEKK4 negatively regulates the autophagic process.


Asunto(s)
Antígenos de Diferenciación/metabolismo , Autofagia , MAP Quinasa Quinasa Quinasa 4/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Antígenos de Diferenciación/genética , Autofagia/efectos de los fármacos , Proteína 5 Relacionada con la Autofagia , Línea Celular , Lipopolisacáridos/toxicidad , Ratones , Proteínas Asociadas a Microtúbulos/genética , Mutagénesis Sitio-Dirigida , Células 3T3 NIH , Fosforilación , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/genética
2.
Oncogene ; 27(50): 6462-72, 2008 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-18955973

RESUMEN

c-MYC has a pivotal function in growth control, differentiation and apoptosis, and its abnormal expression is associated with many tumors. Overexpression of c-MYC sensitizes cells to apoptosis by a variety of stimuli. The decision of a cell to undergo apoptosis and how this apoptotic response is regulated by c-MYC depends on the specific cell type and the physiological status of the cell. Multiple cooperating molecular pathways of cell survival and apoptosis determine whether a cell lives or dies, and understanding how c-MYC interfaces with these pathways to influence the survival of cells is important to understand normal and abnormal development, tumor initiation and progression, and response of tumors to different treatment regimens. This article will provide an overview of the function of the tumor suppressor gene product p53 in the c-MYC-mediated apoptotic response and how c-MYC amplifies the intrinsic mitochondrial pathway and triggers and/or amplifies the death receptor pathways. Finally, a model for how deregulated c-MYC prematurely triggers the normal apoptotic response associated with terminal myeloid differentiation while also blocking the differentiation program is presented.


Asunto(s)
Apoptosis , Genes myc , Transducción de Señal , Animales , Supervivencia Celular , Genes Supresores de Tumor , Humanos , Mitocondrias/metabolismo , Neoplasias/genética , Neoplasias/patología , Proteína p53 Supresora de Tumor/metabolismo
3.
Leukemia ; 22(10): 1909-16, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18650844

RESUMEN

We have shown that deregulated expression of either c-Myb or E2F-1 blocks terminal differentiation of M1 myeloid leukemia cells at the blast stage, whereas deregulated c-Myc blocks differentiation at the intermediate stage. Each of these oncogenes potentiates M1 leukemia in vivo. The zinc-finger transcription factor Egr-1 abrogates the block in M1 terminal differentiation imparted by oncogenic c-Myc or E2F-1, suppressing their leukemia-promoting function in nude mice. In this study, we asked whether Egr-1 also abrogates the block in terminal differentiation and suppresses leukemia imparted by deregulated c-Myb. Interestingly, the ectopic expression of Egr-1 in M1 cells expressing deregulated c-Myb only partially abrogated the block in terminal differentiation and did not suppress the leukemic phenotype. Two important implications from these data are that the leukemia suppressor function of Egr-1 is not directly related to how early the transforming oncogene blocks the differentiation program and that the tumor suppressor function of Egr-1 is dependent on the specific oncogene. Egr-1 is dominant to c-Myc- and E2F-1-, but not to c-Myb-, driven leukemia. These findings extend the notion that the molecular nature of genetic lesions responsible for leukemia determines the effectiveness of any given tumor suppressor.


Asunto(s)
Proteína 1 de la Respuesta de Crecimiento Precoz/fisiología , Genes myb/fisiología , Genes myc/fisiología , Leucemia Mieloide/prevención & control , Proteínas Supresoras de Tumor/fisiología , Animales , Apoptosis , Ciclo Celular , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/análisis , Interleucina-6/farmacología , Leucemia Mieloide/patología , Ratones , Fagocitosis , Fosfotransferasas/análisis , Fosfotransferasas/fisiología
4.
Oncogene ; 27(1): 98-106, 2008 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-17599039

RESUMEN

Deregulated growth and blocks in differentiation collaborate in the multistage process of leukemogenesis. Previously, we have shown that ectopic expression of the zinc finger transcription factor Egr-1 in M1 myeloblastic leukemia cells promotes terminal differentiation with interleukin-6 (IL-6). In addition, we have shown that deregulated expression of the oncogene E2F-1 blocks the myeloid terminal differentiation program, resulting in proliferation of immature cells in the presence of IL-6. Here it is shown that the positive regulator of differentiation Egr-1 abrogates the E2F-1-driven block in myeloid terminal differentiation. The M1E2F-1/Egr-1 cells underwent G(0)/G(1) arrest and functional macrophage maturation following treatment with IL-6. Furthermore, Egr-1 diminished the aggressiveness of M1E2F-1 leukemias and abrogated the leukemic potential of IL-6-treated M1E2F-1 cells. Previously, we reported that Egr-1 abrogated the block in terminal myeloid differentiation imparted by deregulated c-myc, which blocks differentiation at a later stage than E2F-1, resulting in cells that have the characteristics of functionally mature macrophages that did not undergo G(0)/G(1) arrest. Taken together, this work extends and highlights the tumor suppressor role of Egr-1, with Egr-1 behaving as a tumor suppressor against two oncogenes, each blocking myeloid differentiation by a different mechanism. These findings suggest that Egr-1 and/or Egr-1 target genes may be useful tools to treat or suppress oncogene-driven hematological malignancies.


Asunto(s)
Diferenciación Celular/fisiología , Proteína 1 de la Respuesta de Crecimiento Precoz/fisiología , Inhibidores de Crecimiento/fisiología , Leucemia Mieloide Aguda/patología , Leucemia Mieloide Aguda/prevención & control , Células Mieloides/metabolismo , Células Mieloides/patología , Proteínas Supresoras de Tumor/fisiología , Animales , Apoptosis/fisiología , Línea Celular Tumoral , Factor de Transcripción E2F1/antagonistas & inhibidores , Factor de Transcripción E2F1/fisiología , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Inhibidores de Crecimiento/genética , Interleucina-6/fisiología , Leucemia Mieloide Aguda/metabolismo , Macrófagos/citología , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Ratones Desnudos , Células Mieloides/citología
5.
Oncogene ; 26(21): 3041-50, 2007 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-17099722

RESUMEN

M1 myeloid leukemic cells were used to dissect the molecular mechanisms of myeloid cell survival and apoptosis. A salient feature of M1 cells is that they respond to the physiological survival factor interleukin-6 (IL-6), yet lack the tumor suppressor gene p53. Functional wild-type activation of temperature-sensitive p53 protein (p53 val) at permissive temperature in M1-t-p53 cells results in rapid apoptosis, which is blocked by IL-6. How p53 induces M1 apoptosis and how IL-6 protects against p53-induced apoptosis are not fully understood. Here it is shown that p53-mediated apoptosis of M1 cells involves rapid activation of the proapoptotic Fas/CD95 death pathway, which activates caspases 8 and 10. Functional p53 also targets the mitochondria, causing upregulation of proapoptotic Bax, downregulation of prosurvival Bcl-2 and activation of caspase 9. IL-6 was found to protect against p53-induced apoptosis via activation of the PI3K/Akt survival pathway, which in turn counters both the Fas/CD95 and mitochondrial apoptotic pathways and activates the prosurvival transcription factor nuclear factor-kappaB (NF-kappaB). Taken together, this work supports a novel model for leukemic progression where cells that acquire the ability to produce an autocrine survival factor, such as IL-6, can bypass normal p53 surveillance function by targeting Akt, which in turn can exert effects on the regulators of apoptosis, such as the Fas/CD95 pathway, the mitochondria and NF-kappaB.


Asunto(s)
Apoptosis/fisiología , Interleucina-6/fisiología , Leucemia Mieloide/enzimología , Leucemia Mieloide/patología , Fosfatidilinositol 3-Quinasas/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología , Transducción de Señal/fisiología , Proteína p53 Supresora de Tumor/fisiología , Animales , Línea Celular Tumoral , Humanos , Ratones
6.
Oncogene ; 25(40): 5537-46, 2006 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-16732331

RESUMEN

The gadd45 family of gene(s) is rapidly induced by genotoxic stress or by differentiation-inducing cytokines. Using bone marrow (BM) from gadd45a-/-, gadd45b-/- and wild-type (wt) mice, we investigated their role in stress responses of myeloid cells to acute stimulation with differentiating cytokines, myelotoxic agents and inflammatory substances. Bone marrow cells from gadd45a-/- and gadd45b-/- mice displayed compromised myeloid differentiation and higher apoptosis in vitro, following acute stimulation with a variety of differentiating cytokines. Intriguingly, gadd45a-/- and gadd45b-/- colony forming units granulocyte/macrophage progenitors displayed prolonged proliferation capacity compared to wt controls upon re-plating in methylcellulose supplemented with interleukin-3. The recovery of the BM myeloid compartment following 5-Fluorouracil-induced myelo-ablation was much slower in gadd45a-/- and gadd45b-/- mice compared to wt controls. Furthermore, the response of myeloid cells to inflammatory stress, inflicted via intraperitoneal administration of sodium caseinate was impaired in gadd45a-/- and gadd45b-/- mice compared to age-matched wt mice, as indicated by lower percentage of Gr-1-positive cells in the BM and lower number of myeloid cells in peritoneal exudates. Overall, these data indicate that both gadd45a and gadd45b play a role in modulating physiological stress responses of myeloid cells to acute stimulation with differentiating cytokines, myelo-ablation and inflammation. These findings should aid in understanding the response of normal and malignant hematopoietic cells to physiological and chemical stressors including anticancer agents.


Asunto(s)
Antígenos de Diferenciación/fisiología , Proteínas de Ciclo Celular/fisiología , Citocinas/farmacología , Inflamación/fisiopatología , Agonistas Mieloablativos/farmacología , Células Mieloides/fisiología , Proteínas Nucleares/fisiología , Animales , Antígenos de Diferenciación/genética , Apoptosis , Northern Blotting , Células de la Médula Ósea/citología , Caseínas/farmacología , Proteínas de Ciclo Celular/genética , Diferenciación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Femenino , Fémur/citología , Fluorouracilo/farmacología , Células Madre Hematopoyéticas/metabolismo , Ratones , Células Mieloides/citología , Mielopoyesis , Proteínas Nucleares/genética
7.
Leukemia ; 16(4): 527-41, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11960329

RESUMEN

Myeloid differentiation (MyD) primary response and growth arrest DNA damage (Gadd) genes comprise a set of overlapping genes, including known (IRF-1, EGR-1, Jun) and novel (MyD88, Gadd45alpha, MyD118/Gadd45beta, GADD45gamma, MyD116/ Gadd34) genes, that have been cloned by virtue of being co-ordinately induced upon the onset of terminal myeloid differentiation and following exposure of cells to stress stimuli. In recent years it has become evident that MyD/Gadd play a role in blood cell development, where they function as positive regulators of terminal differentiation, lineage-specific blood cell development and control of blood cell homeostasis, including growth inhibition and apoptosis. MyD/Gadd are also involved in inflammatory responses to invading micro-organisms, and response to environmental stress and physiological stress, such as hypoxia, which results in ischemic tissue damage. An intricate network of interactions among MyD/GADD genes and gene products appears to control their diverse functions. Deregulated growth, increased cell survival, compromised differentiation and deficiencies in DNA repair are hallmarks of malignancy and its progression. Thus, the role MyD/Gadd play in negative growth control, including cell cycle arrest and apoptosis, and in DNA repair, make them attractive molecular targets for tumor suppression. The role MyD/Gadd play in innate immunity and host response to hypoxia also make these genes and gene products attractive molecular targets to treat immunity and inflammation disorders, such as septic shock and ischemic tissue damage.


Asunto(s)
Antígenos de Diferenciación/genética , Daño del ADN/genética , Inflamación/terapia , Neoplasias/terapia , Animales , Apoptosis , Ciclo Celular , Terapia Genética , Humanos , Inmunidad , Neoplasias/inmunología
8.
DNA Cell Biol ; 20(4): 239-47, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11403721

RESUMEN

The MyD118 (Gadd45beta) protein is a member of a family of structurally related proteins, including Gadd45 (Gadd45alpha) and CR6 (Gadd45gamma), that have critical roles in regulating growth arrest and apoptosis. The MyD118 and other members of its family display distinct patterns of expression in response to stimuli that induce differentiation, growth arrest, or apoptosis. Species-blot analysis showed that MyD118 is an evolutionarily conserved gene, and comparative sequence analysis showed that MyD118 has a gene structure similar to that of other members of its gene family. Comparison of putative transcription factor-binding sites found in sequences of this gene family provides evidence that p53 is involved in regulating the expression of MyD118 and that NF-kappaB may play a role in differential expression of MyD118 and Gadd45(Gadd45alpha). Fluorescence in situ hybridization localized the MyD118 gene to mouse chromosome band 10B5.3, correcting a previous assignment to mouse chromosome 9.


Asunto(s)
Antígenos de Diferenciación/genética , Genes , Ratones/genética , Secuencia de Aminoácidos , Animales , Antígenos de Diferenciación/fisiología , Secuencia de Bases , Sitios de Unión , Pollos/genética , Mapeo Cromosómico , Cromosomas Humanos Par 19/genética , Perros , Evolución Molecular , Humanos , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Polimorfismo Conformacional Retorcido-Simple , Conejos , Alineación de Secuencia , Homología de Secuencia , Factores de Transcripción/metabolismo
9.
Int J Oncol ; 18(4): 749-57, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11251170

RESUMEN

The MyD118/Gadd45/CR6 gene family (also termed Gadd45beta/alpha/gamma) has been identified as genes which are rapidly induced by genotoxic agents, during terminal differentiation, as well as by apoptotic cytokines. In recent years, evidence has emerged that the proteins encoded by these genes play pivotal roles in negative growth control, including growth suppression and apoptotic cell death. However, under what physiological condition these proteins mediate either cell cycle arrest or apoptosis, and the molecular nature of apoptotic pathways involved are currently unclear. Thus, to further explore the effects of these genes on cell growth and cell viability, either in the presence or absence of extrinsic stress, we have established M1 myeloblastic leukemia and H1299 lung carcinoma cell lines, where high level ectopic expression of MyD118, Gadd45, or CR6 can be induced by isopropyl beta-D-thiogalactopyranoside (IPTG). By taking advantage of these cell lines, it was observed that in the absence of genotoxic stress, inducible expression of MyD118, Gadd45 and/or CR6 resulted in retardation of cellular proliferation and accumulation of cells in the G1 phase of the cell cycle. Ectopic expression of these proteins also was found to sensitize the cells to apoptosis induced by genotoxic agents such as UV, MMS, gamma-irradiation and VP16. Finally, evidence has been obtained that in the absence of stress, ectopic expression of MyD118/Gadd45/CR6 is insufficient to activate the MTKl/JNK/p38 stress cascade, and that enhancement of genotoxic stress induced apoptosis by these proteins may involve apoptotic pathways other than the JNK/p38 pathways.


Asunto(s)
Antígenos de Diferenciación/biosíntesis , Apoptosis , Proteínas Portadoras/biosíntesis , Leucemia Mieloide Aguda/metabolismo , Neoplasias Pulmonares/metabolismo , Estrés Oxidativo , Biosíntesis de Proteínas , Antígenos de Diferenciación/genética , Northern Blotting , Western Blotting , Proteínas Portadoras/genética , Ciclo Celular , Daño del ADN , Reparación del ADN , Etopósido/farmacología , Citometría de Flujo , Regulación de la Expresión Génica/efectos de los fármacos , Vectores Genéticos , Humanos , Péptidos y Proteínas de Señalización Intracelular , Isopropil Tiogalactósido/farmacología , Leucemia Mieloide Aguda/patología , Neoplasias Pulmonares/patología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Proteínas/genética , Transfección , Células Tumorales Cultivadas/efectos de los fármacos , Proteinas GADD45
10.
Blood ; 97(5): 1298-305, 2001 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-11222373

RESUMEN

Using a variety of differentiation-inducible myeloid cell lines, we previously showed that the zinc-finger transcription factor early growth response gene 1 (Egr-1) is a positive modulator of macrophage differentiation and negatively regulates granulocytic differentiation. In this study, high-efficiency retroviral transduction was used to ectopically express Egr-1 in myeloid-enriched or stem cell-enriched bone marrow cultures to explore its effect on the development of hematopoietic progenitors in vitro and in lethally irradiated mice. It was found that ectopic Egr-1 expression in normal hematopoietic progenitors stimulates development along the macrophage lineage at the expense of development along the granulocyte or erythroid lineages, regardless of the cytokine used. Moreover, Egr-1 accelerated macrophage development by suppressing the proliferative phase of the growth-to-macrophage developmental program. The remarkable ability of Egr-1 to dictate macrophage development at the expense of development along other lineages resulted in failure of Egr-1-infected hematopoietic progenitors to repopulate the bone marrow and spleen, and thereby prevent death, in lethally irradiated mice. These observations further highlight the role Egr-1 plays in monocytic differentiation and growth suppression.


Asunto(s)
Proteínas de Unión al ADN/farmacología , Hematopoyesis/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de los fármacos , Proteínas Inmediatas-Precoces , Macrófagos/efectos de los fármacos , Factores de Transcripción/farmacología , Animales , Células de la Médula Ósea/citología , Trasplante de Médula Ósea , Técnicas de Cultivo de Célula , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Proteínas de Unión al ADN/genética , Proteína 1 de la Respuesta de Crecimiento Precoz , Eritrocitos/citología , Femenino , Granulocitos/citología , Macrófagos/citología , Ratones , Ratones Endogámicos BALB C , Factores de Transcripción/genética , Transducción Genética , Irradiación Corporal Total/mortalidad
11.
J Biol Chem ; 276(4): 2766-74, 2001 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-11022036

RESUMEN

GADD45, MyD118, and CR6 (also termed GADD45alpha, beta, and gamma) comprise a family of genes that encode for related proteins playing important roles in negative growth control, including growth suppression. Data accumulated suggest that MyD118/GADD45/CR6 serve similar but not identical functions along different apoptotic and growth suppressive pathways. It is also apparent that individual members of the MyD118/GADD45/CR6 family are differentially induced by a variety of genetic and environmental stress agents. The MyD118, CR6, and GADD45 proteins were shown to predominantly localize within the cell nucleus. Recently, we have shown that both MyD118 and GADD45 interact with proliferating cell nuclear antigen (PCNA), a protein that plays a central role in DNA replication, DNA repair, and cell cycle progression, as well as with the universal cyclin-dependent kinase inhibitor p21. In this work we show that also CR6 interacts with PCNA and p21. Moreover, it is shown that CR6 interacts with PCNA via a domain that also mediates interaction of both GADD45 and MyD118 with PCNA. Importantly, evidence has been obtained that interaction of CR6 with PCNA impedes the function of this protein in negative growth control, similar to observations reported for MyD118 and GADD45.


Asunto(s)
Carcinoma/patología , Proteínas Portadoras/metabolismo , Ciclinas/metabolismo , Neoplasias Pulmonares/patología , Antígeno Nuclear de Célula en Proliferación/metabolismo , Ensayo de Tumor de Célula Madre , Antígenos de Diferenciación , Apoptosis , Sitios de Unión , División Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Humanos , Unión Proteica , Células Tumorales Cultivadas , Técnicas del Sistema de Dos Híbridos
12.
J Cell Biochem ; 79(2): 249-60, 2000 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-10967552

RESUMEN

We have partially purified and characterized the 5-methylcytosine removing activity (5-meC-DNA Glycosylase) from HeLa cells with 700-fold enrichment. This activity cleaves DNA specifically at fully methylated CpG sites. The mechanism of 5-meC removal is base excision from fully methylated CpG loci on DNA, producing abasic sites. Hemi-methylated DNA is not a substrate. A prominent 52 KDa protein is present in all partially purified fractions. This activity is tightly associated with other nuclear factors and proteins, which resulted in differential fractionation of this activity on ion exchange columns. One nuclear factor associated with this activity is identified as RNA. Another nuclear protein, proliferating cell nuclear antigen (PCNA) is also associated with this enzyme. Glycosylic removal of 5-meC from DNA by this activity could be involved in the regulation of transcription, replication, differentiation, and development through resultant hypomethylation of DNA.


Asunto(s)
N-Glicosil Hidrolasas/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , ARN/metabolismo , Secuencia de Bases , Islas de CpG , ADN Glicosilasas , Metilación de ADN , ADN de Cadena Simple , Células HeLa , Humanos , Cinética , Especificidad por Sustrato
13.
Blood ; 96(2): 475-82, 2000 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-10887108

RESUMEN

The transcription factor E2F-1 has been postulated to play a crucial role in the control of cell cycle progression because of its ability to be bound and regulated by the retinoblastoma gene product (pRb). Exogenous expression of E2F-1, under growth restrictive conditions, was shown to result in p53-dependent programmed cell death. The consequences of deregulated expression of E2F-1 on terminal differentiation of hematopoietic cells in the absence of E2F-1-mediated apoptosis, as well as mechanistic insights into how deregulated E2F-1 may affect terminal differentiation, have not been established. The autonomously proliferating M1 myeloblastic leukemia cell line, which is null for p53 expression and can be induced by interleukin-6 (IL-6) to undergo terminal macrophage differentiation with concomitant loss of leukemogenicity, provides a particularly attractive model system to address these issues. Deregulated and continued expression of E2F-1 blocked the IL-6-induced terminal differentiation program at an early blast stage, giving rise to immature cells, which continued to proliferate without undergoing apoptosis and retained their leukemogenic phenotype. Although E2F-1 blocked IL-6-mediated terminal differentiation and its associated growth arrest, it did not prevent the rapid induction of both p15(INK4B) and p16(INK4A), inhibition of cdk4 kinase activity, and subsequent hypophosphorylation of pRb. The results obtained imply that genetic alterations that both impair p53 function and deregulate E2F-1 expression may render hematopoietic cells refractory to the induction of differentiation and are, thereby, likely to play a major role in the progression of leukemias. (Blood. 2000;96:475-482)


Asunto(s)
Proteínas Portadoras/biosíntesis , Proteínas de Ciclo Celular , Diferenciación Celular/efectos de los fármacos , Proteínas de Unión al ADN , Regulación de la Expresión Génica , Leucemia Mieloide Aguda/patología , Factores de Transcripción/farmacología , Proteínas Supresoras de Tumor , Animales , Apoptosis/efectos de los fármacos , División Celular/efectos de los fármacos , Inhibidor p15 de las Quinasas Dependientes de la Ciclina , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Factores de Transcripción E2F , Factor de Transcripción E2F1 , Interleucina-6/farmacología , Leucemia Mieloide Aguda/metabolismo , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Proteínas Recombinantes/farmacología , Proteína 1 de Unión a Retinoblastoma , Factor de Transcripción DP1 , Factores de Transcripción/genética , Células Tumorales Cultivadas
14.
Oncogene ; 19(20): 2447-54, 2000 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-10828887

RESUMEN

Members of the cdc25 family are protein phosphatases that play pivotal roles in cell cycle progression. Cdc25A has been shown to be a critical regulator of the G1/S transition of mammalian cells and to be a myc-target gene with oncongenic properties. We investigated the regulation of cdc25A during terminal differentiation using myeloblastic leukemia M1 cells, that can be induced to undergo differentiation into macrophages by interleukin-6 (IL-6) treatment. In this report it is shown that cdc25A protein is degraded by the ubiquitin-proteasome machinery in both terminally differentiating and cycling cells. Cdc25A was found to have two major peaks of accumulation during cell cycle progression, one in G1 and the other in S/G2. Evidence was obtained that degradation of cdc25A by the ubiquitin-proteasome machinery in terminally differentiating myeloid cells is accelerated compared to cycling cells. Moreover, deregulated expression of c-myc in M1 cells, which had been previously shown to block terminal differentiation, was also found to block IL-6 induced degradation of cdc25A.


Asunto(s)
Ciclo Celular , Diferenciación Celular , Cisteína Endopeptidasas/metabolismo , Complejos Multienzimáticos/metabolismo , Ubiquitinas/metabolismo , Fosfatasas cdc25/metabolismo , Complejo de la Endopetidasa Proteasomal , ARN Mensajero/genética , ARN Mensajero/metabolismo , Fosfatasas cdc25/genética
15.
Oncogene ; 19(26): 2967-77, 2000 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-10871848

RESUMEN

Previously we have shown that deregulated expression of c-myc in M1 myeloid leukemic cells blocked IL-6-induced differentiation and its associated growth arrest; however, the cells proliferated at a significantly reduced rate compared to untreated cells. The basis for the increased doubling time of IL-6-treated M1myc cells was found to be due to the induction of a p53-independent apoptotic pathway. The apoptotic response was not completely penetrant; in the same population of cells both proliferation and apoptosis were continuously ongoing. Down-regulation of Bcl-2 was insufficient to account for the apoptotic response, since deregulated expression of Bcl-2 delayed, but did not block, the onset of apoptosis. Furthermore, our results indicated that the IL-6-induced partial hypophosphorylation of the retinoblastoma gene product (Rb), observed in M1myc cells, was not responsible for the apoptotic response. Finally, the findings in M1 cells were extended to myeloid cells derived from the bone marrow of wild type and p53-deficient mice, where the deregulated expression of c-myc was also shown to block terminal differentiation and induce apoptosis independent of p53. These findings provide new insights into how myc participates in the neoplastic process, and how additional mutations can promote more aggressive tumors. Oncogene (2000) 19, 2967 - 2977


Asunto(s)
Apoptosis/fisiología , Proteínas de Ciclo Celular , Diferenciación Celular/fisiología , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Leucemia Mieloide Aguda/patología , Proteínas Proto-Oncogénicas c-myc/fisiología , Proteína p53 Supresora de Tumor/fisiología , Proteínas Supresoras de Tumor , Animales , Médula Ósea/metabolismo , Médula Ósea/patología , Proteínas Portadoras/fisiología , Inhibidor p15 de las Quinasas Dependientes de la Ciclina , Humanos , Interleucina-6/farmacología , Ratones , Ratones Endogámicos BALB C , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteína de Retinoblastoma/fisiología
16.
J Biol Chem ; 275(22): 16810-9, 2000 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-10828065

RESUMEN

MyD118 and Gadd45 are related genes encoding for proteins that play important roles in negative growth control, including growth suppression and apoptosis. MyD118 and Gadd45 are related proteins that previously were shown to interact with proliferating cell nuclear antigen (PCNA), implicated in DNA replication, DNA repair, and cell cycle progression. To establish the role of MyD118 and Gadd45 interactions with PCNA, in this work we sought to identify the interacting domains and analyze the significance of this interaction in negative growth control. Using complementary in vivo and in vitro interaction assays the N-terminal (1-46) and middle (100-127) regions of PCNA were identified as harboring MyD118- and Gadd45 interacting domains, whereas PCNA interacting domains within MyD118 and Gadd45 were localized to the C termini of these proteins (amino acids 114-156 and 137-165, respectively). These findings provide first evidence that similar domains within MyD118 and Gadd45 mediate interactions with PCNA. Importantly, ectopic expression of MyD118 or Gadd45 N-terminal peptides, lacking the PCNA interacting domain, was found to suppress colony formation or induce apoptosis more efficiently than the full-length proteins. These findings suggest that interaction of MyD118 or Gadd45 with PCNA, in essence, serves to impede negative growth control.


Asunto(s)
Antígenos de Diferenciación/metabolismo , División Celular , Antígeno Nuclear de Célula en Proliferación/metabolismo , Proteínas/metabolismo , Antígenos de Diferenciación/química , Apoptosis , Sitios de Unión , Línea Celular , Ensayo de Inmunoadsorción Enzimática , Humanos , Péptidos y Proteínas de Señalización Intracelular , Antígeno Nuclear de Célula en Proliferación/química , Proteínas/antagonistas & inhibidores , Proteínas/química , Técnicas del Sistema de Dos Híbridos , Proteinas GADD45
17.
Oncogene ; 18(35): 4899-907, 1999 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-10490824

RESUMEN

MyD118 and Gadd45 are two related genes which encode for proteins that play important roles in negative growth control, including both growth suppression and apoptosis. A strategy was employed to clone new members of the MyD118 and Gadd45 family of genes. Based on alignment of the deduced amino acid sequences, one cDNA clone was found to encode for the murine homologue of human CR6, originally cloned as an IL-2 immediate-early response gene. The murine and human CR6 proteins were observed to be 97% identical, indicating that CR6 is an evolutionarily conserved protein. Analysis of CR6 expression during hematopoietic cell development associated with growth arrest and apoptotic cell death, upon exposure of hematopoietic cells to a variety of growth arrest and apoptotic stimuli, and in a variety of murine tissues, has revealed that CR6 expression differs significantly from the expression of the related MyD118 and Gadd45 genes. Nevertheless, CR6, like MyD118 and Gadd45, suppressed colony formation of human lung carcinoma H1299 cells. These data suggest that CR6 plays similar, but not identical, roles to MyD118 and Gadd45 in negative control of cell growth.


Asunto(s)
Antígenos de Diferenciación/genética , Apoptosis , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Hematopoyesis , Péptidos y Proteínas de Señalización Intracelular , Proteínas/genética , Secuencia de Aminoácidos , Animales , Antígenos de Diferenciación/química , Antígenos de Diferenciación/metabolismo , Apoptosis/efectos de los fármacos , Apoptosis/genética , Secuencia de Bases , Proteínas Sanguíneas/farmacología , Proteínas Portadoras/química , Diferenciación Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Línea Celular , Clonación Molecular , Secuencia Conservada/genética , Regulación de la Expresión Génica/efectos de los fármacos , Sustancias de Crecimiento/farmacología , Hematopoyesis/efectos de los fármacos , Humanos , Metilmetanosulfonato/farmacología , Ratones , Datos de Secuencia Molecular , Mutágenos/farmacología , Proteínas/química , Proteínas/metabolismo , ARN Mensajero/análisis , ARN Mensajero/genética , Células Tumorales Cultivadas , Proteinas GADD45
18.
J Biol Chem ; 274(40): 28697-707, 1999 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-10497240

RESUMEN

Interleukin (IL)-6 plays an important role in a wide range of biological activities, including differentiation of murine M1 myeloid leukemic cells into mature macrophages. At the onset of M1 differentiation, a set of myeloid differentiation primary response (MyD) genes are induced, including the proto-oncogene for JunB. In order to examine the molecular nature of the mechanisms by which IL-6 activates the immediate early expression of MyD genes, JunB was used as a paradigm. A novel IL-6 response element, -65/-52 IL-6RE, to which a 100-kDa protein complex is bound, has been identified on the JunB promoter. Leukemia inhibitory factor (LIF)-induced activation of JunB in M1 cells was also mediated via the -65/-52 IL-6RE. The STAT3 and CRE-like binding sites of the JunB promoter, identified as IL-6-responsive elements in HepG2 liver cells were found, however, to play no role in JunB inducibility by IL-6 in M1 myeloid cells. Conversely, the -65/-52 IL-6RE is shown not to be necessary for JunB inducibility by IL-6 or LIF in liver cells. It appears, therefore, that immediate early activation of JunB is regulated differently in M1 myeloid cells than in HepG2 liver cells. This indicates that distinct cis-acting control elements participate in cell type-specific induction of JunB by members of the IL-6 cytokine superfamily.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Inhibidores de Crecimiento/farmacología , Interleucina-6/farmacología , Linfocinas/farmacología , Proteínas Proto-Oncogénicas c-jun/genética , Animales , Secuencia de Bases , Sitios de Unión , ADN , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica/genética , Factor Inhibidor de Leucemia , Ratones , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , Factor de Transcripción STAT3 , Transactivadores/metabolismo , Células Tumorales Cultivadas
19.
Oncogene ; 17(10): 1189-94, 1998 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-9771961

RESUMEN

Meeting's Report -- June 2, 1998, Sugarload Estate Conference Center, Philadelphia, Pennsylvania, USA. A symposium on Normal Development, Oncogenesis and Programmed Cell Death, was held at the Sugarload Estate Conference Center, Philadelphia, Pennsylvania, USA sponsored by the Fels Cancer Institute, Temple University School of Medicine, with the support of the Alliance Pharmaceutical Corporation. The symposium was organized by Drs Dan A Liebermann and Barbara Hoffman at the Fels. Invited speakers included: Dr Andrei V Gudkov (University of Illinois) who started the symposium talking about 'New cellular factors modulating the tumor suppressor function of p53'; Dr Yuri Lazebnik (Cold Spring Harbor Laboratories) spoke about 'Caspases considered as enemies within'; Dr E Premkumar Reddy (Fels Institute, Temple University) talked about recent exciting findings in his laboratory regarding 'JAK-STATs dedicated signaling pathways'; Dr Michael Greenberg (Harvard University) spoke about 'Signal transduction pathways that regulate differentiation and survival in the developing nervous system'; Dr Richard Kolesnick's (Memorial Sloan-Kettering Cancer Center) talk has been focused at 'Stress signals for apoptosis, including Ceramide and c-Jun Kinase/Stress-activated Protein Kinase'; Dr Barbara Hoffman (Fels Institute, Temple University) described research, conducted in collaboration with Dr Dan A Liebermann, aimed at deciphering the roles of 'myc, myb, and E2F as negative regulators of terminal differentiation', using hematopoietic cells as model system. Dr Daniel G Tenen (Harvard Medical School), described studies aimed at understanding the 'Regulation of hematopoietic cell development by lineage specific transcription regulators'. Dr George C Prendergast (The Wistar Institute) talked about the 'Myc-Bin1 signaling pathway in cell death and differentiation. Dr Ruth J Muschel (University of Pennsylvania) spoke about work, conducted in collaboration with Dr WG McKenna, aimed at gaining a better understanding of 'Radioresistance and the cell cycle'. Finally Dr Donald Kufe concluded the symposium (Dana Farber Cancer Institute, Harvard Medical School) describing studies that were performed in his laboratory addressing the 'Role for the c-Abl tyrosine kinase in genetic recombination'.


Asunto(s)
Apoptosis/genética , Fenómenos Fisiológicos Celulares , Neoplasias/etiología , Animales , Ciclo Celular , Diferenciación Celular , Humanos , Oncogenes , Transducción de Señal
20.
Blood ; 92(6): 1957-66, 1998 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-9731053

RESUMEN

We previously have shown that the zinc finger transcription factor Egr-1 blocked granulocytic differentiation of HL-60 cells, restricting differentiation along the monocytic lineage. Egr-1 also was observed to block granulocyte colony-stimulating factor (G-CSF)-induced differentiation of interleukin-3 (IL-3)-dependent 32Dcl3 hematopoietic precursor cells, endowing the cells with the ability to be induced by granulocyte-macrophage colony-stimulating factor (GM-CSF) for terminal differentiation along the macrophage lineage. To better understand the function of Egr-1 as a positive modulator of monocytic differentiation, in this work we have studied the effect of ectopic expression of Egr-1 on the murine myeloblastic leukemic cell line M1, which is induced for differentiation by the physiological inducer IL-6. It is shown that, unlike in HL-60 and 32Dcl3 cells, ectopic expression of Egr-1 in M1 cells resulted in activation of the macrophage differentiation program in the absence of differentiation inducer. This included the appearance of morphologically differentiated cells, decreased growth rate in mass culture, and cloning efficiency in soft agar, and expression of endogenous c-myb and c-myc mRNAs was markedly downregulated. Untreated M1Egr-1 cells also exhibited cell adherence, expression of Fc and C3 receptors, and upregulation of the myeloid differentiation primary response genes c-Jun, junD, and junB and the late genetic markers ferritin light-chain and lysozyme. Ectopic expression of Egr-1 in M1 cells also dramatically increased the sensitivity of the cells for IL-6-induced differentiation, allowed a higher proportion of M1 cells to become terminally differentiated under conditions of optimal stimulation for differentiation, and decreased M1 leukemogenicity in vivo. These findings demonstrate that the functions of Egr-1 as a positive modulator of macrophage differentiation vary, depending on the state of lineage commitment for differentiation of the hematopoietic cell type.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Proteínas Inmediatas-Precoces , Leucemia Mieloide Aguda/patología , Macrófagos/patología , Factores de Transcripción/fisiología , Dedos de Zinc/fisiología , Animales , Diferenciación Celular/efectos de los fármacos , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/genética , Proteína 1 de la Respuesta de Crecimiento Precoz , Regulación Neoplásica de la Expresión Génica/genética , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Interleucina-6/biosíntesis , Leucemia Mieloide Aguda/etiología , Leucemia Mieloide Aguda/metabolismo , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Receptores de Interleucina-6/biosíntesis , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética , Transgenes/genética , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA